TW201836642A - Methods of treating autoimmune and inflammatory diseases - Google Patents

Methods of treating autoimmune and inflammatory diseases Download PDF

Info

Publication number
TW201836642A
TW201836642A TW107110152A TW107110152A TW201836642A TW 201836642 A TW201836642 A TW 201836642A TW 107110152 A TW107110152 A TW 107110152A TW 107110152 A TW107110152 A TW 107110152A TW 201836642 A TW201836642 A TW 201836642A
Authority
TW
Taiwan
Prior art keywords
content
individual
sample
analysis
biomarkers
Prior art date
Application number
TW107110152A
Other languages
Chinese (zh)
Inventor
麥可 約翰 湯森
傑森 駭克尼
南帝尼 拉馬穆斯
Original Assignee
美商建南德克公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 美商建南德克公司 filed Critical 美商建南德克公司
Publication of TW201836642A publication Critical patent/TW201836642A/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/564Immunoassay; Biospecific binding assay; Materials therefor for pre-existing immune complex or autoimmune disease, i.e. systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, rheumatoid factors or complement components C1-C9
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/24Immunology or allergic disorders
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Analytical Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Public Health (AREA)
  • Biophysics (AREA)
  • Veterinary Medicine (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Rehabilitation Therapy (AREA)
  • Rheumatology (AREA)
  • Cell Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Provided herein are biomarkers and therapies for the treatment of autoimmune and/or inflammatory diseases, such as lupus, and methods of using BTK inhibitors. In particular, provided are biomarkers for patient selection and prognosis in lupus, as well as methods of therapeutic treatment, articles of manufacture and methods for making them, diagnostic kits, methods of detection and methods of advertising related thereto.

Description

治療自體免疫及發炎疾病的方法Methods for treating autoimmune and inflammatory diseases

本文提供用於治療自體免疫及發炎疾病之生物標記物及療法,以及使用BTK抑制劑之方法。特定而言,提供用於選擇自體免疫及發炎疾病患者及預後之生物標記物,以及醫療性治療之方法、製品及其製造方法、診斷套組、偵測方法以及與其相關之推廣方法。Provided herein are biomarkers and therapies for treating autoimmune and inflammatory diseases, as well as methods of using BTK inhibitors. In particular, it provides methods for selecting patients with autoimmune and inflammatory diseases and prognosis, as well as medical treatment methods, products and manufacturing methods, diagnostic kits, detection methods, and related promotion methods.

自體免疫及發炎疾病以及病症仍然顯著威脅人類健康。儘管在治療自體免疫及發炎疾病以及病症方面已顯著進步,但仍尋求改良療法。許多自體免疫及發炎疾病展現不均一性跡象。舉例而言,全身性紅斑性狼瘡症(systemic lupus erythematosus;SLE)為在SLE患者群中具有不均一性跡象之疾病。參見Kennedy等人, Lupus Sci. & Med., 2015; 2:e000080。鑒於此不均一性,除治療自體免疫及發炎疾病(例如,SLE)之新方法以外,存在對使用可改良治療結果之診斷生物標記物鑑別某些患者之方法的需求。 漿母細胞為快速分裂、短生命期抗體分泌細胞。在幼年型狼瘡患者血液中已鑑別出漿母細胞之增加,且總體而言增加狼瘡患者中之抗體轉錄物之豐度。E.Arce等人,J. Immunol. 167 , 2361-2369 (2001);L, Bennett等人,J. Exp. Med. 197 , 711-723 (2003)。雖然漿母細胞表示血液中之較小比例之B細胞,但其負責全血mRNA中發現之大部分抗體轉錄物。 蛋白質激酶為人類酶之最大家族,其涵蓋遠遠超過500種蛋白質。布魯頓氏酪胺酸激酶(Bruton's Tyrosine Kinase;BTK)為酪胺酸激酶之Tec家族之成員,且為早期B細胞發育以及成熟B細胞活化、信號傳導及存活之調節子。BTK缺陷型小鼠模型中已確定BTK在過敏性病症及/或自體免疫疾病及/或發炎疾病方面之作用的跡象。舉例而言,在標準鼠類SLE臨床前模型中,BTK缺陷已展示使得顯著減緩疾病進展。此外,BTK缺陷型小鼠亦對患上膠原蛋白誘導之關節炎具有抗性且可能較不易患葡萄球菌(Staphylococcus)誘導之關節炎。大量證據支持B細胞及體液免疫系統在自體免疫及/或發炎疾病之發病機制中之作用。參見例如WO 2012/118750。發展至消耗B細胞之基於蛋白質之治療劑(諸如美羅華(Rituxan))表示治療多種自體免疫及/或發炎疾病之方法。由於BTK在B細胞活化中之作用,BTK抑制劑可適用作B細胞介導之病原性活性(諸如自體抗體生產)之抑制劑。BTK亦在破骨細胞(osteoclasts)、肥大細胞及單核球中表現且已展示對於此等細胞之功能而言為重要的。舉例而言,小鼠之BTK缺乏症與IgE介導之肥大細胞活化受損(TNF-α及其他發炎性細胞介素釋放顯著減弱)相關,且人類之BTK缺乏症與經活化單核球之TNF-α生產大大減少相關。 抑制BTK活性可適用於治療過敏性病症及/或自體免疫及/或發炎疾病,諸如:SLE、類風濕性關節炎、多血管炎、特發性血小板減少性紫癜(ITP)、重症肌無力、過敏性鼻炎及哮喘(Di Paolo等人(2011) Nature Chem. Biol. 7(1):41-50;Liu等人(2011) Jour.of Pharm.及Exper. Ther. 338(1):154-163)。已報導特定BTK抑制劑(Liu (2011) Drug Metab.及Disposition 39(10):1840-1849;美國專利第7,884,108號;WO 2010/056875;美國專利第7,405,295號;美國專利第7,393,848號;WO 2006/053121;美國專利第7,947,835號;US 2008/0139557;美國專利第7,838,523號;US 2008/0125417;US 2011/0118233;2011年8月31日提交之PCT/US2011/050034 「PYRIDINONES/PYRAZINONES, METHOD OF MAKING, AND METHOD OF USE THEREOF」;2011年8月31日提交之PCT/US2011/050013 「PYRIDAZINONES, METHOD OF MAKING, AND METHOD OF USE THEREOF」;2011年5月6日提交之美國序列第13/102,720號「PYRIDONE AND AZA - PYRIDONE COMPOUNDS AND METHODS OF USE」)。 美國專利第8,716,274號(以全文引用之方式併入本文中)揭示適用於抑制BTK之雜芳基吡啶及氮雜吡啶酮化合物之類別。下文描繪之化合物(A)為一種特定BTK抑制劑化合物:。 化合物(A)為:(S)-2-(3'-(羥基甲基)-1-甲基-5-((5-(2-甲基-4-(氧雜環丁-3-基)哌嗪-1-基)吡啶-2-基)胺基)-6-側氧基-1,6-二氫-[3,4'-二吡啶]-2'-基)-7,7-二甲基-2,3,4,6,7,8-六氫-1H-環戊[4,5]吡咯并[1,2-a]吡嗪-1-酮。在化學結構與化學名稱之間存在任何不一致性之情況下,化學結構佔優勢。 本文所引用之所有參考文獻,包括專利申請案及公開案,以全文引用之方式併入本文中。Autoimmune and inflammatory diseases and conditions remain a significant threat to human health. Despite significant advances in treating autoimmune and inflammatory diseases and conditions, improved therapies are still sought. Many autoimmune and inflammatory diseases show signs of heterogeneity. For example, systemic lupus erythematosus (SLE) is a disease that shows signs of heterogeneity in a population of SLE patients. See Kennedy et al., Lupus Sci. & Med., 2015; 2: e000080. In view of this heterogeneity, in addition to new methods for treating autoimmune and inflammatory diseases (e.g., SLE), there is a need for a method to identify certain patients using diagnostic biomarkers that can improve treatment outcomes. Plasmablasts are fast-dividing, short-lived antibody-secreting cells. Increases in plasmablasts have been identified in the blood of juvenile lupus patients, and overall increase the abundance of antibody transcripts in lupus patients. E. Arce et al., J. Immunol. 167 , 2361-2369 (2001); L, Bennett et al., J. Exp. Med. 197 , 711-723 (2003). Although plasmablasts represent a small percentage of B cells in the blood, they are responsible for most antibody transcripts found in whole blood mRNAs. Protein kinases are the largest family of human enzymes, covering far more than 500 proteins. Bruton's Tyrosine Kinase (BTK) is a member of the Tec family of tyrosine kinases and is a regulator of early B cell development and mature B cell activation, signaling, and survival. Signs of the role of BTK in allergic and / or autoimmune and / or inflammatory diseases have been identified in BTK-deficient mouse models. For example, in a standard preclinical murine SLE model, BTK defects have been shown to significantly slow disease progression. In addition, BTK-deficient mice are also resistant to collagen-induced arthritis and may be less susceptible to Staphylococcus-induced arthritis. Substantial evidence supports the role of B cells and the humoral immune system in the pathogenesis of autoimmune and / or inflammatory diseases. See, for example, WO 2012/118750. Protein-based therapeutics that develop to deplete B cells, such as Rituxan, represent a method of treating a variety of autoimmune and / or inflammatory diseases. Because of the role of BTK in B cell activation, BTK inhibitors are suitable as inhibitors of B cell-mediated pathogenic activities, such as autoantibody production. BTK has also been shown in osteoclasts, mast cells and monocytes and has been shown to be important for the function of these cells. For example, BTK deficiency in mice is associated with impaired IgE-mediated mast cell activation (significantly reduced release of TNF-α and other inflammatory interleukins), and BTK deficiency in humans is associated with activated monocytes. TNF-α production is significantly reduced. Inhibition of BTK activity is suitable for the treatment of allergic conditions and / or autoimmune and / or inflammatory diseases such as: SLE, rheumatoid arthritis, polyangiitis, idiopathic thrombocytopenic purpura (ITP), myasthenia gravis Allergic rhinitis and asthma (Di Paolo et al. (2011) Nature Chem. Biol. 7 (1): 41-50; Liu et al. (2011) Jour. Of Pharm. And Exper. Ther. 338 (1): 154 -163). Specific BTK inhibitors have been reported (Liu (2011) Drug Metab. And Disposition 39 (10): 1840-1849; US Patent No. 7,884,108; WO 2010/056875; US Patent No. 7,405,295; US Patent No. 7,393,848; WO 2006 / 053121; US Patent No. 7,947,835; US 2008/0139557; US Patent No. 7,838,523; US 2008/0125417; US 2011/0118233; PCT / US2011 / 050034 "PYRIDINONES / PYRAZINONES, METHOD OF" filed on August 31, 2011 MAKING, AND METHOD OF USE THEREOF "; PCT / US2011 / 050013" PYRIDAZINONES, METHOD OF MAKING, AND METHOD OF USE THEREOF "filed on August 31, 2011; US Serial No. 13 / 102,720 filed on May 6, 2011 "PYRIDONE AND AZA-PYRIDONE COMPOUNDS AND METHODS OF USE"). U.S. Patent No. 8,716,274, which is incorporated herein by reference in its entirety, discloses classes of heteroarylpyridine and azapyridone compounds suitable for inhibiting BTK. Compound (A) depicted below is a specific BTK inhibitor compound: . Compound (A) is: (S) -2- (3 '-(hydroxymethyl) -1-methyl-5-((5- (2-methyl-4- (oxetan-3-yl ) Piperazin-1-yl) pyridin-2-yl) amino) -6- pendantoxy-1,6-dihydro- [3,4'-dipyridine] -2'-yl) -7,7 -Dimethyl-2,3,4,6,7,8-hexahydro-1H-cyclopenta [4,5] pyrrolo [1,2-a] pyrazin-1-one. In the case of any inconsistency between the chemical structure and the chemical name, the chemical structure prevails. All references cited herein, including patent applications and publications, are incorporated herein by reference in their entirety.

本文提供一種用於治療患有自體免疫或發炎疾病之個體的方法,該方法包含向該個體投與治療有效量之BTK抑制劑,其中已發現來自個體之樣本具有升高含量之選自由IgJ、Mzb1及Txndc5組成之群的一或多種生物標記物。 本文亦提供一種用於治療個體之自體免疫或發炎疾病的方法,該方法包含: (a)測定來自個體之樣本包含升高含量之選自由IgJ、Mzb1及Txndc5組成之群的一或多種生物標記物;以及 (b)向個體投與有效量之BTK抑制劑,藉此治療免疫疾病或病症。 本文亦提供一種用於選擇用於患有自體免疫或發炎疾病之個體之療法的方法,該方法包含測定選自由IgJ、Mzb1及Txndc5組成之群的一或多種生物標記物的含量;以及基於生物標記物之含量選擇藥物。 本文亦提供一種鑑別或多或少有可能展現受益於包含BTK抑制劑之治療的患有自體免疫或發炎疾病之個體的方法,其係藉由測定來自個體之樣本中之選自由IgJ、Mzb1及Txndc5組成之群的一或多種生物標記物的含量,其中樣本中之升高含量之生物標記物指示個體更有可能展現受益於包含BTK抑制劑之治療,或降低含量之生物標記物指示個體不大可能展現受益於包含BTK抑制劑之治療。 本文亦提供一種用於鑑別接受BTK抑制劑之患有自體免疫或發炎疾病之個體的分析法,該方法包含: (a)測定來自個體之樣本中之選自由IgJ、Mzb1及Txndc5組成之群的一或多種生物標記物之含量;以及 (b)基於生物標記物之含量而推薦投與BTK抑制劑。 本文亦提供一種包含一或多種反應劑之診斷套組,其用於測定來自患有自體免疫或發炎疾病之個體之樣本中之選自由IgJ、Mzb1及Txndc5組成之群的一或多種生物標記物的含量,其中偵測到升高含量之生物標記物意謂當用BTK抑制劑治療個體時療效提高,且其中偵測到較低或大體上不可偵測含量之生物標記物意謂當用BTK抑制劑治療患有自體免疫或發炎疾病之個體時療效降低。 在本文提供之方法之一些實施例中,該方法進一步包含向個體投與有效量之BTK抑制劑。 在本文提供之方法、分析法及/或套組之一些實施例中,樣本為血液樣本。 在本文提供之方法、分析法及/或套組之一些實施例中,BTK抑制劑為抗體、結合多肽、小分子及/或聚核苷酸。 在本文提供之方法、分析法及/或套組之一些實施例中,BTK抑制劑為小分子。在一些實施例中,小分子BTK抑制劑為化合物(A)或其醫藥學上可接受之鹽。 在本文提供之方法、分析法及/或套組之一些實施例中,自體免疫或發炎疾病為全身性紅斑性狼瘡症。在一些實施例中,自體免疫或發炎疾病為狼瘡性腎炎。在一些實施例中,自體免疫或發炎疾病為腎外狼瘡。 先前已揭示用於預測自體免疫疾病(諸如類風濕性關節炎、多發性硬化症及狼瘡)中對經B細胞拮抗劑(例如,抗CD20抗體)治療之反應的生物標記物物及其使用方法,但不為本發明漿母細胞基因標籤,且不關於BTK抑制。參見WO 2012/118750,其全部內容以引用之方式併入本文中。 如本文所提供,B細胞子集之轉錄分析鑑別出對漿母細胞具有特異性之基因表現標籤。在實驗中,此標籤在實驗中與體外尖峰中之漿母細胞豐度高度相關。對SLE患者中之B細胞子集使用FACS分析,與RNA定序配對,本發明基因表現標籤展示與全血中之漿母細胞之頻率的較強相關性。雖然漿母細胞表示血液中之較小比例之B細胞,但其負責全血mRNA中發現之大部分抗體轉錄物。擴展至兩個額外II期臨床試驗群,使用SLEDAI疾病活動指數發現漿母細胞標籤與疾病活動性相關。在存在抗DNA抗體、較低含量之補體及白血球減少症之情況下,此關聯係由漿母細胞之相關性驅動。增加之漿母細胞標籤亦與高含量之干擾素活性相關聯。與疾病嚴重程度無關,患者人種/種族亦預示漿母細胞標籤含量。標準護理藥劑,特定言之黴酚酸酯,減少漿母細胞標記基因之表現。舉例而言,藉由用利妥昔單抗(rituximab)治療患者引起漿母細胞標籤表現之大幅(但最終短暫的)減少。Provided herein is a method for treating an individual suffering from an autoimmune or inflammatory disease, the method comprising administering to the individual a therapeutically effective amount of a BTK inhibitor, wherein a sample from the individual has been found to have an elevated content selected from IgJ , Mzb1 and Txndc5 in one or more biomarkers. Also provided herein is a method for treating an autoimmune or inflammatory disease in an individual, the method comprising: (a) determining that a sample from the individual comprises an elevated content of one or more organisms selected from the group consisting of IgJ, Mzb1, and Txndc5 A marker; and (b) administering to a subject an effective amount of a BTK inhibitor, thereby treating an immune disease or disorder. Also provided herein is a method for selecting a therapy for an individual suffering from an autoimmune or inflammatory disease, the method comprising determining the content of one or more biomarkers selected from the group consisting of IgJ, Mzb1, and Txndc5; and based on The content of the biomarker is selected as the drug. This article also provides a method for identifying individuals with autoimmune or inflammatory diseases that are more or less likely to exhibit benefit from treatments comprising BTK inhibitors by measuring a sample from an individual selected from IgJ, Mzb1 The content of one or more biomarkers in a group consisting of Txndc5 and Txndc5, where an increased amount of the biomarker in the sample indicates that the individual is more likely to exhibit benefit from a treatment that includes a BTK inhibitor, or a reduced content of the biomarker indicates the individual It is unlikely to show benefit from a treatment comprising a BTK inhibitor. Also provided herein is an assay for identifying an individual with an autoimmune or inflammatory disease receiving a BTK inhibitor, the method comprising: (a) determining a sample from the individual selected from the group consisting of IgJ, Mzb1, and Txndc5 Content of one or more biomarkers; and (b) it is recommended to administer a BTK inhibitor based on the biomarker content. Also provided herein is a diagnostic kit comprising one or more reagents for determining one or more biomarkers selected from the group consisting of IgJ, Mzb1, and Txndc5 in a sample from an individual suffering from an autoimmune or inflammatory disease. Biomarkers in which elevated levels of biomarkers are detected mean improved efficacy when the subject is treated with a BTK inhibitor, and in which lower or substantially undetectable levels of biomarkers are meant to be used BTK inhibitors are less effective in treating individuals with autoimmune or inflammatory diseases. In some embodiments of the methods provided herein, the method further comprises administering to the individual an effective amount of a BTK inhibitor. In some embodiments of the methods, assays and / or kits provided herein, the sample is a blood sample. In some embodiments of the methods, assays and / or kits provided herein, the BTK inhibitor is an antibody, binding polypeptide, small molecule, and / or polynucleotide. In some embodiments of the methods, assays and / or kits provided herein, the BTK inhibitor is a small molecule. In some embodiments, the small molecule BTK inhibitor is Compound (A) or a pharmaceutically acceptable salt thereof. In some embodiments of the methods, assays and / or kits provided herein, the autoimmune or inflammatory disease is systemic lupus erythematosus. In some embodiments, the autoimmune or inflammatory disease is lupus nephritis. In some embodiments, the autoimmune or inflammatory disease is extrarenal lupus. Biomarkers and their uses for predicting response to treatment with B cell antagonists (e.g., anti-CD20 antibodies) in autoimmune diseases such as rheumatoid arthritis, multiple sclerosis, and lupus have been previously disclosed The method, but not the plasmablast gene tag of the present invention, is not related to BTK inhibition. See WO 2012/118750, the entire contents of which are incorporated herein by reference. As provided herein, transcriptional analysis of a subset of B cells identifies genetic expression tags specific to plasmablasts. In experiments, this label was highly correlated with plasmablast abundance in spikes in vitro. Using FACS analysis on a subset of B cells in SLE patients, paired with RNA sequencing, the gene expression tag of the present invention shows a strong correlation with the frequency of plasmablasts in whole blood. Although plasmablasts represent a small percentage of B cells in the blood, they are responsible for most antibody transcripts found in whole blood mRNAs. Expansion into two additional Phase II clinical trials using the SLEDAI disease activity index to find plasmablast labeling associated with disease activity. In the presence of anti-DNA antibodies, lower levels of complement and leukocytopenia, this relationship is driven by the correlation of plasmablasts. Increased plasmablast labeling is also associated with high levels of interferon activity. Regardless of the severity of the disease, the patient's race / ethnicity also predicts plasmablast labeling. Standard care agents, specifically mycophenolate mofetil, reduce the performance of plasmablast marker genes. For example, treating patients with rituximab causes a significant (but ultimately transient) reduction in plasmablast labeling.

I. 定義 如在本文中可互換地使用,「聚核苷酸」或「核酸」係指任何長度之核苷酸之聚合物且包括DNA及RNA。核苷酸可為脫氧核糖核苷酸、核糖核苷酸、經修飾之核苷酸或鹼基及/或其類似物,或可藉由DNA或RNA聚合酶或藉由合成反應併入聚合物中之任何受質。聚核苷酸可包含經修飾核苷酸,諸如甲基化核苷酸及其類似物。若存在,則可在聚合物組裝之前或之後賦予對核苷酸結構之修飾。核苷酸序列可間雜有非核苷酸組分。聚核苷酸可在合成之後進一步修飾,諸如與標記結合。其他類型之修飾包括例如「封端」;用類似物取代一或多種天然產生之核苷酸;核苷酸間修飾,諸如具有不帶電鍵聯(例如,膦酸甲酯、磷酸三酯、磷醯胺、胺基甲酸酯等)及具有帶電鍵聯(例如,硫代磷酸酯、二硫代磷酸酯等)之彼等、含有諸如蛋白質(例如,核酸酶、毒素、抗體、訊號肽、聚-L-離胺酸等)之側接部分之彼等、具有嵌入劑(例如,吖啶、補骨脂素等)之彼等、含有螯合劑(例如,金屬、放射性金屬、硼、氧化金屬等)之彼等、含有烷基化劑之彼等、具有經修飾鍵聯(例如,α變旋異構核酸等)之彼等,以及聚核苷酸之未經修飾形式。此外,一般存在於糖中之任何羥基可例如由膦酸酯基、磷酸酯基置換,由標準保護基保護,或經活化以製備與額外核苷酸之額外鍵聯,或可結合至固體或半固體支撐物。5'及3'末端OH可經磷酸化或經1至20個碳原子之胺或有機封端基團部分取代。其他羥基亦可衍生成標準保護基。聚核苷酸亦可含有此項技術中通常已知的類似形式之核糖或脫氧核糖,包括例如2'-O-甲基-、2'-O-烯丙基、2'-氟-或2'-疊氮基-核糖、碳環糖類似物、α-變旋異構糖、差向異構糖(諸如阿拉伯糖(arabinose)、木糖(xyloses)或來蘇糖(lyxoses));哌喃醣、呋喃醣、景天庚糖(sedoheptuloses)、非環類似物及無鹼基核苷類似物(諸如甲基核糖苷)。一或多個磷酸二酯鍵可經替代性鍵聯基團置換。此等替代性鍵聯基團包括(但不限於)其中磷酸經P(O)S(「硫代酸酯」)、P(S)S (「二硫代酸酯」)、(O)NR2 (「醯胺化物」)、P(O)R、P(O)OR'、CO或CH2 (「甲縮醛」)置換之實施例,其中各R或R'獨立地為H或經視情況含有醚(-O-)鍵、芳基、烯基、環烷基、環烯基或芳烷基之經取代或未經取代之烷基(1-20 C)。聚核苷酸中並非所有鍵需要一致。前述描述適用於本文所提及之所有聚核苷酸,包括RNA及DNA。如本文中所使用,「寡核苷酸」一般係指較短單股聚核苷酸,但長度不必小於250個核苷酸。寡核苷酸可為合成的。術語「寡核苷酸」及「聚核苷酸」並非相互排斥。以上關於聚核苷酸之描述同樣且完全適用於寡核苷酸。術語「引子」係指通常藉由提供游離3'-OH基團能夠與核酸雜交,且隨後與互補核酸聚合之單股聚核苷酸。術語「小分子」係指分子量為約2000道爾頓或更小,較佳約500道爾頓或更小之任何分子。 術語「宿主細胞」、「宿主細胞株」及「宿主細胞培養物」可互換地使用且係指已引入外源核酸之細胞,包括此等細胞之後代。宿主細胞包括「轉型體」及「轉型細胞」,其包括初級轉型細胞及自其衍生之後代且不考慮繼代數。後代之核酸含量與母細胞可能不完全相同,但可能含有突變。本文包括針對原始轉型細胞篩檢或選擇具有相同功能或生物活性之突變型後代。 如本文中所使用,術語「載體」係指一種核酸分子,其能夠傳送其所連接之另一核酸。該術語包括作為自我複製核酸結構之載體以及併入已引入其之宿主細胞之基因組中的載體。某些載體能夠導引可操作地連結其之核酸的表現。此等載體在本文中稱為「表現載體」。 「經分離」抗體為已與其天然環境之組分分離的抗體。在一些實施例中,如藉由例如電泳(例如,SDS-PAGE、等電聚焦(IEF)、毛細電泳法)或層析(例如,離子交換或逆相HPLC)所測定,將抗體純化為至於95%或99%純度。關於評估抗體純度之方法的綜述,參見例如Flatman等人,J. Chromatogr. B 848:79-87 (2007)。 「經分離」核酸係指已與其天然環境之組分分離的核酸分子。經分離核酸包括通常含有核酸分子之細胞中所含的核酸分子,但該核酸分子存在於染色體外或在不同於其天然染色體位置之染色體位置處。 術語「抗體」在本文中以最廣泛意義使用且涵蓋各種抗體結構,包括(但不限於)單株抗體、多株抗體、多特異性抗體(例如,雙特異性抗體)及抗體片段,只要其展現所需抗原結合活性即可。 「阻斷」抗體或「拮抗劑」抗體為抑制或降低其所結合之抗原之生物活性的抗體。較佳阻斷抗體或拮抗劑抗體實質上或完全抑制抗原之生物活性。 「親和力」係指分子(例如,抗體)之單一結合位點與其結合搭配物(例如,抗原)之間的非共價相互作用之總和的強度。除非另外指示,否則如本文中所使用,「結合親和力」係指反映結合對(例如,抗體與抗原)之成員之間的1:1相互作用的固有結合親和力。分子X對其搭配物Y之親和力一般可由解離常數(Kd)表示。可藉由此項技術中已知之常用方法(包括本文中所描述的彼等方法)來量測親和力。用於量測結合親和力之特定說明性及例示性實施例描述於下文中。 「親和力成熟」抗體係指相較於在一或多個高變區(HVR)中不具有一或多個變化之親本抗體,在一或多個高變區中具有此等變化之抗體,此等變化使抗體對抗原之親和力得到改良。 術語「偵測」包括任何偵測方式,包括直接及間接偵測。 如本文中所使用,術語「生物標記物」係指可在樣本中偵測到之指示物,例如預測性、診斷性及/或預後性。生物標記物可充當由某些分子、病理性、組織學及/或臨床特徵之疾病或病症(例如,癌症)表徵之特定次型的指示物。在一些實施例中,生物標記物為基因。生物標記物包括(但不限於)聚核苷酸(例如,DNA及/或RNA)、多肽、多肽及聚核苷酸修飾(例如,轉譯後修飾)、碳水化合物及/或糖脂類分子標記物。 術語「生物標記物標籤」、「標籤」、「生物標記物表現標籤」或「表現標籤」在本文中可互換地使用且係指一種生物標記物或其組合,該生物標記物之表現為指示物,例如預測性、診斷性及/或預後性。生物標記物標籤可充當由某些分子、病理性、組織學及/或臨床特徵之疾病或病症(例如,癌症)表徵之特定次型的指示物。在一些實施例中,生物標記物標籤為「基因標籤」。術語「基因標籤」可與「基因表現標籤」互換地使用且係指一種聚核苷酸或其組合,該聚核苷酸之表現為指示物,例如預測性、診斷性及/或預後性。在一些實施例中,生物標記物標籤為「蛋白質標籤」。術語「蛋白質標籤」可與「蛋白質表現標籤」互換地使用且係指一種多肽或其組合,該多肽之表現為指示物,例如預測性、診斷性及/或預後性。 與對個體之增加的臨床益處相關的生物標記物之「量」或「含量」為樣本中之可偵測含量。此等可藉由熟習此項技術者已知且本文亦揭示之方法量測。所評估之生物標記物之表現含量或量可用於確定對治療之反應。 術語「表現之含量」或「表現含量」一般可互換地使用且一般係指生物樣本中之生物標記物的量。「表現」一般係指藉由其將資訊(例如,基因編碼及/或表觀遺傳)轉換成存在於細胞中且在細胞中操作之結構的方法。因此,如本文中所使用,「表現」可指轉錄成聚核苷酸、轉譯成多肽或甚至聚核苷酸及/或多肽修飾(例如,多肽之轉譯後修飾)。轉錄聚核苷酸、轉譯多肽或聚核苷酸及/或多肽修飾(例如,多肽之轉譯後修飾)之片段亦應被視為表現,無論其來源於由替代性剪接產生之轉錄物或降解之轉錄物,還是來源於例如藉由蛋白分解之多肽轉譯後加工。「表現基因」包括轉錄成如mRNA之聚核苷酸,且隨後轉譯成多肽之基因,且亦包括轉錄成RNA,但未轉譯成多肽(例如,轉移及核糖體RNA)之基因。 「高表現」、「高表現含量」或「高含量」係指個體中之生物標記物相對於對照物(諸如個體或未患疾病或病症(例如,癌症)之個體或內部對照(例如,管家生物標記物))之增加的表現或增加的含量。 「降低表現」、「降低表現含量」或「降低含量」係指個體中之生物標記物相對於對照物(諸如個體或未患疾病或病症(例如,癌症)之個體或內部對照(例如,管家生物標記物))之減少的表現或減少的含量。在一些實施例中,減少的表現為極少表現或無表現。 在某些實施例中,術語「在參考含量下」係指基本上等同於參考含量的來自個體或患者之樣本中之生物標記物的含量或係指與參考含量相差高達1%、高達2%、高達3%、高達4%、高達5%的含量。在一些實施例中,參考含量為參考群體中之生物標記物的中間含量。在一些實施例中,標記物之參考含量為參考群體中之標記物的平均含量。在一些實施例中,標記物之參考含量為參考群體中之標記物之平均含量。 在某些實施例中,術語「高於參考含量」係指相較於參考含量,藉由本文所描述之方法測定,高於參考含量至少5%、10%、20%、25%、30%、40%、50%、60%、70%、80%、85%、90%、95%、100%或更大的來自個體或患者之樣本中之生物標記物的含量。在一些實施例中,參考含量為參考群體中之中間含量。在一些實施例中,標記物之參考含量為參考群體中之標記物的平均含量。 在某些實施例中,術語「低於參考含量」係指相較於參考含量,藉由本文所描述之方法測定,低於參考含量至少5%、10%、20%、25%、30%、40%、50%、60%、70%、80%、85%、90%、95%、100%或更大的來自個體或患者之樣本中之生物標記物的含量。在一些實施例中,參考含量為參考群體中之中間含量。在一些實施例中,標記物之參考含量為參考群體中之標記物的平均含量。在一些實施例中,標記物之參考含量為參考群體中之標記物之平均含量。 術語「管家生物標記物」係指通常類似地存在於所有細胞類型中之生物標記物或生物標記物群(例如,聚核苷酸及/或多肽)。在一些實施例中,管家生物標記物為「管家基因」。「管家基因」在本文中係指編碼對於細胞功能維持來說不可或缺且通常類似地存在於所有細胞類型中之蛋白質的基因或基因群。 如本文中所使用,「擴增」一般係指產生所需序列之多個複本的方法。「多個複本」意謂至少兩個複本。「複本」不一定意謂與模板序列互補或一致之完美序列。舉例而言,複本可包括核苷酸類似物,諸如脫氧肌苷、有意序列改變(諸如經由包含與模板可雜交但不互補之序列之引子引入的序列改變),及/或在擴增期間發生之序列誤差。 術語「多重PCR」係指出於擴增單個反應中之兩個或更多個DNA序列之目的而使用超過一個引子在自單個來源(例如,個體)獲得之核酸上進行之單個PCR反應。 雜交反應之「嚴格度」可易於由一般熟習此項技術者測定,且通常為視探針長度、洗滌溫度及鹽濃度而定之經驗計算。大體而言,愈長探針需要愈高溫度以用於適當退火,而較短探針需要低溫。當互補股存在於低於其熔融溫度之環境中時,雜交通常視變性DNA再退火之能力而定。探針與可雜交序列之間的所需同源性度愈高,其可使用之相對溫度愈高。因此,由此可見愈高相對溫度將傾向於使反應條件更加嚴格,同時溫度亦降低。雜交反應之嚴格度的額外細節及解釋參見Ausubel等人, Current Protocols in Molecular Biology, Wiley Interscience Publishers, (1995)。 如本文中所定義,「嚴格條件」或「高嚴格度條件」可藉由以下來鑑別:(1)採用低離子強度及高溫以用於洗滌,例如在50℃下,0.015 M氯化鈉/0.0015 M檸檬酸鈉/0.1%十二烷基硫酸鈉;(2)在雜交期間,採用變性試劑,諸如甲醯胺,例如在42℃下,具有0.1%牛血清白蛋白/0.1%菲科爾(Ficoll)/0.1%聚乙烯吡咯啶酮/pH 6.5之50mM磷酸鈉緩衝劑以及750 mM氯化鈉、75 mM檸檬酸鈉之50%(v/v)甲醯胺;或(3)在42℃下在採用50%甲醯胺、5 × SSC (0.75 M NaCl、0.075 M檸檬酸鈉)、50 mM磷酸鈉(pH 6.8)、0.1%焦磷酸鈉、5 × 鄧哈特溶液(Denhardt's solution)、超聲處理鮭魚精子DNA (50 μg/ml)、0.1%SDS及10%硫酸葡聚糖之溶液中雜交隔夜,伴以在42℃下在0.2 × SSC (氯化鈉/檸檬酸鈉)中之10分鐘洗滌,隨後在55℃下進行由含有EDTA之0.1 × SSC組成之10分鐘高嚴格度洗滌。 「中等嚴格條件」可如由Sambrook等人, Molecular Cloning :A Laboratory Manual,New York:Cold Spring Harbor Press,1989所描述來鑑別,且包括使用洗滌溶液及比上文所描述的彼等更寬鬆之雜交條件(例如,溫度、離子強度及%SDS)。中等嚴格條件之實例為在37℃下在包含以下之溶液中培育隔夜:20%甲醯胺、5 × SSC (150 mM NaCl、15 mM檸檬酸三鈉)、50 mM磷酸鈉(pH 7.6)、5×鄧哈特溶液、10%硫酸葡聚糖及20 mg/ml變性剪切鮭魚精子DNA,隨後在約37-50℃下在1×SSC中洗滌過濾物。熟習此項技術者將知道如何視需要調節溫度、離子強度等以適應諸如探針長度及其類似物的因素。 術語「診斷」在本文中用以指代分子或病理學病況、疾病或病狀(例如,癌症)之鑑別或分類。舉例而言,「診斷」可指特定類型癌症之鑑別。「診斷」亦可指例如藉由組織病理學標準,或藉由分子特徵(例如,由一種生物標記物 (例如,由該基因編碼之特定基因或蛋白質)之或其組合之表現表徵的亞型)將癌症特定亞型進行分類。 術語「輔助診斷」在本文中係指輔助作出關於疾病或病症(例如,癌症)之特定類型之症狀或病狀之存在或性質之臨床測定的方法。例如,輔助診斷疾病或病狀(例如,癌症)之方法可包括量測來自個體之生物樣本中之某些生物標記物。 如本文中所使用,術語「樣本」係指獲自或衍生自所關注受試者及/或個體的組合物,其含有例如可依據物理、生物化學、化學及/或生理特徵判別及/或鑑定之細胞及/或其他分子實體。舉例而言,片語「疾病樣本」及其變型係指獲自所關注受試者之任何樣本,其預期或已知含有待判別特徵之細胞及/或分子實體。樣本包括(但不限於)原生或培養之細胞或細胞株、細胞上清液、細胞溶胞物、血小板、血清、血漿、玻璃狀液、淋巴液、滑液、濾泡液、精液、羊膜液、乳汁、全血、源自血液之細胞、尿、腦脊髓液、唾液、痰、淚液、汗液、黏液、腫瘤溶胞物及組織培養介質、組織提取物(諸如均質化組織)、腫瘤組織、細胞提取物及其組合。 「組織樣本」或「細胞樣本」意謂獲自受試者或個體之組織之類似細胞的集合。組織或細胞樣本之來源可為固體組織,如:來自新鮮、冷凍及/或保存之器官、組織樣本、活檢體及/或抽出物;血液或任何血液成分,諸如血漿;體液,諸如腦脊髓液、羊膜液、腹膜液或間質液;來自受試者之妊娠或發育之任何時間的細胞。組織樣本亦可為原生或培養之細胞或細胞株。視情況,組織或細胞樣本獲自疾病組織/器官。組織樣本可含有不會與自然界之組織天然互混之化合物,諸如防腐劑、抗凝血劑、緩衝劑、固定劑、營養素、抗生素或其類似物。 如本文中所使用,「參考樣本」、「參考細胞」、「參考組織」、「對照樣本」、「對照細胞」或「對照組織」係指用於達成比較目的之樣本、細胞、組織、標準或含量。在一個實施例中,參考樣本、參考細胞、參考組織、對照樣本、對照細胞或對照組織獲自同一位受試者或個體之身體之健康及/或未患病部分(例如,組織或細胞)。舉例而言,靠近患病細胞或組織(例如,靠近腫瘤之細胞或組織)之健康及/或未患病細胞或組織。在另一實施例中,參考樣本獲自同一位受試者或個體身體之未處理組織及/或細胞。在又一實施例中,參考樣本、參考細胞、參考組織、對照樣本、對照細胞或對照組織獲自不為該受試者或個體之另一位個體身體的健康及/或非患病部分(例如,組織或細胞)。在又另一實施例中,參考樣本、參考細胞、參考組織、對照樣本、對照細胞或對照組織獲自不為該受試者或個體之另一位個體身體之未處理組織及/或細胞。 出於本文之目的,組織樣本之「切片」意謂組織樣本之單一部分或碎片,例如自組織樣本切割的組織或細胞之薄片層。應理解,組織樣本之多個切片可經採用並進行分析,其限制條件為應理解組織樣本之相同切片可在形態及分子含量兩者下進行分析,或關於多肽及聚核苷酸兩者進行分析。 「使...相關(correlate/ correlating)」意謂以任何方式將第一分析或方案之性能及/或結果與第二分析或方案之性能及/或結果進行比較。舉例而言,吾人可使用第一分析或方案之結果進行第二方案及/或吾人可使用第一分析或方案之結果來測定是否應執行第二分析或方案。關於聚核苷酸分析或方案之實施例,吾人可使用聚核苷酸表現分析或方案之結果來測定是否應執行特定治療方案。 「個體反應」或「反應」可使用指示對個體有益之任何端點來進行評估,包括(但不限於):(1)在一定程度上抑制疾病進展(例如,癌症進展),包括減慢及完全遏止;(2)減小腫瘤尺寸;(3)抑制(亦即減輕、減慢或完成遏止)癌細胞穿透至靠近周邊器官及/或組織;(4)抑制(亦即,減輕、減慢或完成遏止)病變;(5)在一定程度上緩解與疾病或病症(例如,癌症)相關聯之一或多種症狀;(6)增加無進展存活之長度;及/或(7)在治療後之給定時間點減小死亡率。 如本文中所使用,術語「大體上相同」表示兩個數值之間的足夠高類似度,使得在藉由該等值(例如,Kd值或表現)量測之生物特徵之情形下,熟習此項技術者將認為兩個值之間的差異為極小或無生物及/或統計顯著性。該兩個值之間的差異為例如小於約50%、小於約40%、小於約30%、小於約20%及/或小於約10%,隨參考/比較值而變化。 如本文中所使用,片語「大體上不同」表示兩個數值之間的足夠高差異程度,使得在藉由該等值(例如,Kd值)量測之生物特徵的情形下,熟習此項技術者將認為這兩個值之間的差異具有統計顯著性。該兩個值之間的差異為例如大於約10%、大於約20%、大於約30%、大於約40%及/或大於約50%,隨參考/比較分子之值而變化。 字組「標記」在在本文中使用時係指可偵測化合物或組合物。標記通常直接或間接地結合或融合至反應劑,諸如聚核苷酸探針或抗體,且有助於偵測到其所結合或融合之反應劑。標記自身可為可偵測的(例如,放射性同位素標記或螢光標記),或在酶標記之情況下,可催化受質化合物或組合物之化學改變,產生可偵測產物。 試劑之「有效量」係指在所需劑量及時間段下,可有效達成所需治療或預防結果的量。 物質/分子(促效劑或拮抗劑)之「治療有效量」可根據以下因素改變:諸如個體之疾病病況、年齡、性別及體重,及物質/分子(促效劑或拮抗劑)在個體體內引發所要反應之能力。治療有效量亦為治療學上有利作用超過物質/分子(促效劑或拮抗劑)之任何有毒或有害作用的量。「預防有效量」係指在所需劑量及時間段下,可有效實現所需預防結果的量。由於個體在患病之前或在疾病早期使用預防劑量,因此預防有效量通常(但不一定)小於治療有效量。 術語「醫藥調配物」係指呈准許其中所含活性成分之生物活性有效之形式的製劑,且其不含對調配物將投與之個體具有不可接受毒性之其他組分。 「醫藥學上可接受之載劑」係指醫藥調配物中之除活性成分以外之對個體無毒的成分。醫藥學上可接受之載劑包括(但不限於)緩衝劑、賦形劑、穩定劑或防腐劑。 如本文中所使用,「治療(treatment)」(及其語法變化形式,諸如「治療(treat/treating)」)係指試圖改變所治療個體之自然病程的臨床介入,且可出於防治目的或在臨床病理學之病程期間進行。所需治療作用包括(但不限於)預防疾病發生或復發,緩解症狀,減輕疾病之任何直接或間接病理性結果,預防癌轉移,減緩疾病進展速率,改善或緩和疾病病況及緩解或改良預後。在一些實施例中,抗體用於延緩疾病發展或減緩疾病進展。 如本申請案中所使用,術語「前藥」係指相較於親本藥物,對腫瘤細胞具有較少細胞毒性的醫藥學上活性物質之前驅體或衍生物形式且能夠以酶方式活化或轉換成更活性親本形式。參看例如Wilman, 「Prodrugs in Cancer Chemotherapy」Biochemical Society Transactions , 14, 第375-382頁, 615th Meeting Belfast (1986)及Stella等人, 「Prodrugs: A Chemical Approach to Targeted Drug Delivery」,Directed Drug Delivery , Borchardt等人, (編), 第247-267頁, Humana Press (1985)。本發明之前藥包括(但不限於)含磷酸鹽前藥、含硫代磷酸鹽前藥、含硫酸鹽前藥、含肽前藥、經D-胺基酸修飾之前藥、糖基化前藥、含β-內醯胺前藥、含視情況經取代之苯氧基乙醯胺前藥或含視情況經取代之苯基乙醯胺前藥、5-氟胞嘧啶及可轉換成更活性細胞毒性游離藥物之其他5-氟尿苷前藥。用於本發明之可衍生為前藥形式之細胞毒性藥物的實例包括(但不限於)上文所描述之彼等化學治療劑。 「個體」或「受試者」為哺乳動物。哺乳動物包括(但不限於)家養動物(例如,牛、羊、貓、狗及馬)、靈長類動物(例如,人類及非人類靈長類,諸如猴)、家兔及嚙齒動物(例如,小鼠及大鼠)。在某些實施例中,個體或受試者為人類。 術語「同時」在本文中用以指代投與兩種或更多種治療劑,其中投與之至少一部分在時間上重疊。因此,同時投與包括在中斷投與一或多種其他試劑之後繼續投與一或多種試劑時的給藥方案。 「降低或抑制」意謂使得整體減少20%、30%、40%、50%、60%、70%、75%、80%、85%、90%、95%或更多的能力。降低或抑制可指經治療病症之症狀、癌轉移之存在或大小或原發腫瘤之大小。 術語「藥品說明書」用以指通常包括於治療性產品之商業包裝中的說明書,其含有關於與使用此等治療性產品有關之適應症、用法、劑量、投與、組合療法、禁忌症及/或警告的資訊。 「製品」為包含至少一種反應劑的任何製造品(例如,包裝或容器)或套組,例如用於治療疾病或病症(例如,癌症)之藥物或用於特異性地偵測本文中所描述的生物標記物之探針。在某些實施例中,製造品或套組係以用於執行本文所述方法之單元形式推銷、分銷或出售。 片語「基於」在本文中使用時意謂關於一或多種生物標記之資訊係用於告知提供於藥品說明書或銷售/促銷指南等上之治療決策、資訊。 如熟習此項技術者所瞭解,本文中提及「約」值或參數包括(且描述)針對該值或參數本身之實施例。舉例而言,提及「約X」之描述包括「X」本身之描述。 應理解,本文所述之態樣及實施例包括「組成」及/或「基本上組成為」態樣及實施例。除非另外規定,否則如本文中所使用,單數形式「一(a/an)」及「該(the)」包括複數個參考物。 II. 方法及用途 本文提供利用漿母細胞生物標記物之方法。特定而言,利用BTK抑制劑及漿母細胞生物標記物之方法。舉例而言,提供用於治療患有疾病或病症之個體的方法,該等方法包含若已發現個體具有漿母細胞生物標記物之存在及/或升高含量,則向該個體投與治療有效量之BTK抑制劑。另外,本文提供用於治療個體之疾病或病症的方法,該方法包含:測定來自個體之樣本包含升高含量之漿母細胞生物標記物,且向該個體投與有效量之BTK抑制劑,藉此治療疾病或病症。在一些實施例中,漿母細胞生物標記物選自由以下基因標籤組成之群:IgJ、Mzb1及Txndc5。在一些實施例中,IgJ、Mzb1及Txndc5之基因表現為多肽表現,其藉由量測患者血液中之該基因之mRNA含量相對於參考含量來測定。在一些實施例中,疾病或病症為自體免疫或發炎疾病或病症。在一些實施例中,疾病或病症為SLE。在一些實施例中,疾病或病症為狼瘡性腎炎。在一些實施例中,疾病或病症為腎外狼瘡。 本文提供治療個體之疾病或病症的方法,該等方法包含向個體投與有效量之BTK抑制劑,其中治療係基於在來自個體之樣本中漿母細胞生物標記物之存在及/或升高含量。在一些實施例中,漿母細胞生物標記物為IgJ、Mzb1及Txndc5中之一或多者之表現。在一些實施例中,IgJ、Mzb1及Txndc5之基因表現為多肽表現,其藉由量測患者血液中之該基因之mRNA含量相對於參考含量來測定。在一些實施例中,疾病或病症為自體免疫或發炎疾病或病症。在一些實施例中,疾病或病症為SLE。在一些實施例中,疾病或病症為狼瘡性腎炎。在一些實施例中,疾病或病症為腎外狼瘡。 此外,本文提供用於選擇用於患有疾病或病症之個體之療法的方法,該等方法包含測定漿母細胞生物標記物之存在及/或含量,且基於生物標記物之存在及/或含量而選擇藥物。在一些實施例中,基於升高含量之漿母細胞生物標記物來選擇藥物。在一些實施例中,漿母細胞生物標記物選自由以下基因標籤組成之群:IgJ、Mzb1及Txndc5。在一些實施例中,IgJ、Mzb1及Txndc5之基因表現為多肽表現,其藉由量測患者血液中之該基因之mRNA含量相對於參考含量來測定。在一些實施例中,疾病或病症為自體免疫或發炎疾病或病症。在一些實施例中,疾病或病症為SLE。在一些實施例中,疾病或病症為狼瘡性腎炎。在一些實施例中,疾病或病症為腎外狼瘡。 本文提供鑑別或多或少有可能展現受益於包含BTK抑制劑之治療的患有疾病或病症之個體的方法,該方法包含:測定來自個體之樣本中之漿母細胞生物標記物之存在及/或含量,其中樣本中之漿母細胞生物標記物之存在及/或升高之含量指示個體更有可能展現受益於包含BTK抑制劑之治療,或漿母細胞生物標記物之缺失及/或降低之含量指示個體不大可能展現受益於包含BTK抑制劑之治療。在一些實施例中,漿母細胞生物標記物選自由以下基因標籤組成之群:IgJ、Mzb1及Txndc5。在一些實施例中,IgJ、Mzb1及Txndc5之基因表現為多肽表現,其藉由量測患者血液中之該基因之mRNA含量相對於參考含量來測定。在一些實施例中,疾病或病症為自體免疫或發炎疾病或病症。在一些實施例中,疾病或病症為SLE。在一些實施例中,疾病或病症為狼瘡性腎炎。在一些實施例中,疾病或病症為腎外狼瘡。 本文亦提供用於鑑別接受BTK抑制劑的患有疾病或病症之個體的分析法,該方法包含:(a)測定來自個體之樣本中之漿母細胞生物標記物之存在及/或含量;(b)基於漿母細胞生物標記物之存在及/或含量而推薦BTK抑制劑。在一些實施例中,基於升高含量之漿母細胞生物標記物來推薦BTK抑制劑。在一些實施例中,漿母細胞生物標記物選自由以下基因標籤組成之群:IgJ、Mzb1及Txndc5。在一些實施例中,IgJ、Mzb1及Txndc5之基因表現為多肽表現,其藉由量測患者血液中之該基因之mRNA含量相對於參考含量來測定。在一些實施例中,疾病或病症為自體免疫或發炎疾病或病症。在一些實施例中,疾病或病症為SLE。在一些實施例中,疾病或病症為狼瘡性腎炎。在一些實施例中,疾病或病症為腎外狼瘡。 本文提供包含一或多種反應劑之診斷套組,其用於測定來自患有疾病或病症之個體之樣本中之漿母細胞生物標記物的含量,其中偵測到漿母細胞生物標記物之存在及/或升高含量意謂當用BTK抑制劑治療個體時療效提高,且其中偵測到較低或大體上不可偵測含量之漿母細胞生物標記物意謂當用BTK抑制劑治療患有疾病之個體時療效降低。本文亦提供製品,該等製品包含包裝在一起之包含BTK抑制劑之醫藥組合物及藥品說明書,該藥品說明書指示BTK抑制劑基於漿母細胞生物標記物之表現而用於治療患有疾病或病症之患者。在一些實施例中,漿母細胞生物標記物選自由以下基因標籤組成之群:IgJ、Mzb1及Txndc5。在一些實施例中,IgJ、Mzb1及Txndc5之基因表現為多肽表現,其藉由量測患者血液中之該基因之mRNA含量相對於參考含量來測定。在一些實施例中,疾病或病症為自體免疫或發炎疾病或病症。在一些實施例中,疾病或病症為SLE。在一些實施例中,疾病或病症為狼瘡性腎炎。在一些實施例中,疾病或病症為腎外狼瘡。 另外,本文提供用於治療個體之疾病或病症的方法,該等方法包含向個體投與有效量之BTK抑制劑,且在用BTK抑制劑治療期間評估來自個體之樣本中之一或多種漿母細胞生物標記物之含量(例如,相較於參考)。亦提供治療個體之疾病或病症的方法,該等方法包含向個體投與有效量之BTK抑制劑,其中治療係基於來自個體之樣本中之一或多種漿母細胞生物標記物之含量(例如,相較於參考)。提供監測包含BTK抑制劑之治療的個體之反應的方法,該方法包含:測定來自個體之樣本中之一或多種漿母細胞生物標記物之含量,其中樣本中之降低含量之一或多種漿母細胞生物標記物(例如,相較於參考)指示個體更有可能回應於包含BTK抑制劑之治療,升高含量及/或與治療前含量大體上相同之含量之一或多種漿母細胞生物標記物(例如,相較於參考)指示個體不大可能回應於包含BTK抑制劑之治療。在一些實施例中,漿母細胞生物標記物選自由以下基因標籤組成之群:IgJ、Mzb1及Txndc5。在一些實施例中,IgJ、Mzb1及Txndc5之基因表現為多肽表現,其藉由量測患者血液中之該基因之mRNA含量相對於參考含量來測定。在一些實施例中,疾病或病症為自體免疫或發炎疾病或病症。在一些實施例中,疾病或病症為SLE。在一些實施例中,疾病或病症為狼瘡性腎炎。在一些實施例中,疾病或病症為腎外狼瘡。 另外提供測定患有疾病或病症之個體是否應繼續或中斷包含BTK抑制劑之治療的方法,該方法包含量測來自個體之樣本中之一或多種漿母細胞生物標記物之含量,其中升高含量及/或與治療前含量大體上相同之含量之一或多種漿母細胞生物標記物(例如,相較於參考)測定個體應中斷包含BTK抑制劑之治療,且降低含量之一或多種漿母細胞生物標記物(例如,相較於參考)測定個體應繼續包含BTK抑制劑之治療。在一些實施例中,漿母細胞生物標記物選自由以下基因標籤組成之群:IgJ、Mzb1及Txndc5。在一些實施例中,IgJ、Mzb1及Txndc5之基因表現為多肽表現,其藉由量測患者血液中之該基因之mRNA含量相對於參考含量來測定。在一些實施例中,疾病或病症為自體免疫或發炎疾病或病症。在一些實施例中,疾病或病症為SLE。在一些實施例中,疾病或病症為狼瘡性腎炎。在一些實施例中,疾病或病症為腎外狼瘡。 在一些實施例中,該方法包含:(a)量測來自患者之生物樣本中之選自IgJ、TXNDC5及MZB1之一種、兩種或三種生物標記物之RNA含量;(b)將(a)中所量測之RNA含量與參考含量進行比較;以及(c)當(a)中所量測之RNA含量高於參考含量時將患者鑑別為更有可能受益於BTK抑制劑療法。在一些實施例中,RNA為mRNA。在一些實施例中,量測mRNA含量包含擴增。在一些實施例中,量測mRNA含量包含定量PCR。在一些實施例中,量測mRNA含量包含擴增mRNA且偵測擴增產物,由此量測mRNA之含量。在一些實施例中,參考含量為參考群體中之對應標記物的中間含量。 在一些實施例中,標記物之參考含量為參考群體中之標記物之中間含量。在本文所描述之實施例中之任一者中,參考含量可為參考群體中之對應標記物之平均含量。在一些實施例中,標記物之參考含量為參考群體中之標記物之平均含量。非限制性例示性參考群體包括患有免疫或發炎疾病之患者、健康個體以及包括健康個體及患有免疫或發炎疾病之患者的群體。在一些實施例中,參考群體包含患有SLE之患者。 在一些實施例中,分析或偵測生物標記物之方法具有小於0.05之p值。在一些實施例中,方法具有高於80%之特異性。在一些實施例中,方法具有高於80%之敏感性。在一些實施例中,方法具有高於70%之ROC。在一些實施例中,方法具有高於70%之AUC。在一些實施例中,方法具有高於70%之正預測值。在一些實施例中,方法具有高於70%之負預測值。在一些實施例中,該參考基因表現概況係來自患者及/或健康志願者之參考群體中之受試者。在一些實施例中,比較步驟包含以下中之至少一者:將表現概況之數位圖像進行比較且將表現資料之資料庫進行比較。 在以上方法中之任一者的一些實施例中,漿母細胞生物標記物為IgJ。在以上方法中之任一者的一些實施例中,漿母細胞生物標記物為Mzb1。在以上方法中之任一者的一些實施例中,漿母細胞生物標記物為Txndc5。在以上方法中之任一者的一些實施例中,一或多種漿母細胞生物標記物為IgJ及Mzb1。在以上方法中之任一者的一些實施例中,一或多種漿母細胞生物標記物為IgJ及Txndc5。在以上方法中之任一者的一些實施例中,一或多種漿母細胞生物標記物為Txndc5及Mzb1。在以上方法中之任一者的一些實施例中,一或多種漿母細胞生物標記物為IgJ、Mzb1及Txndc5。 在以上實施例中之一些中,樣本為尿樣本。在一些實施例中,樣本為血液樣本。在一些實施例中,生物樣本選自血液、血清、血漿及周邊血液單核細胞(PBMC)。在一些實施例中,生物樣本為獲自血液之RNA,例如全血或血液之細胞片段,諸如PBMC。在一些實施例中,生物樣本為血清或血漿。可在治療之前、治療期間或治療後採集樣本。樣本可獲自疑似患有或診斷患有SLE或其他免疫或發炎疾病,且因此有可能需要治療之患者。可替代地,樣本可獲自不疑似患有任何疾病之正常個體。在一些實施例中,在偵測或量測標記物之mRNA含量之前,自本文所描述之生物樣本中提取RNA。 生物標記物之存在及/或表現含量/量可基於此項技術中已知之任何合適的準則而定性及/或定量的測定,包括(但不限於) DNA、mRNA、cDNA、蛋白質、蛋白質片段及/或基因複本數。在某些實施例中,相較於第二樣本中之存在/缺失及/或表現含量/量,第一樣本中之生物標記物之存在及/或表現含量/量增加。在某些實施例中,相較於第二樣本中之存在及/或表現含量/量,第一樣本中之生物標記物之存在/缺失及/或表現含量/量減少。在某些實施例中,第二樣本為參考樣本、參考細胞、參考組織、對照樣本、對照細胞或對照組織。本文描述用於測定基因之存在/缺失及/或表現含量/量之額外揭示內容。 在方法中之任一者的一些實施例中,高表現係指相較於參考樣本、參考細胞、參考組織、對照樣本、對照細胞或對照組織,藉由標準領域已知之方法(諸如本文中所描述的彼等)偵測之生物標記物(例如,蛋白或核酸(例如,基因或mRNA))之含量之10%、20%、30%、40%、50%、60%、70%、80%、90%、95%、96%、97%、98%、99%或更大中之任一者之總體增加。在某些實施例中,高表現係指樣本中之生物標記物之表現含量/量增加,其中增加為參考樣本、參考細胞、參考組織、對照樣本、對照細胞或對照組織中之相應生物標記物之表現含量/量的至少約1.5×、1.75×、2×、3×、4×、5×、6×、7×、8×、9×、10×、25×、50×、75×或100×中之任一者。在一些實施例中,高表現係指相較於參考樣本、參考細胞、參考組織、對照樣本、對照細胞、對照組織或內部對照(例如,管家基因)大約1.5倍、約1.75倍、約2倍、約2.25倍、約2.5倍、約2.75倍、約3.0倍或約3.25倍之總體增加。 在方法中之任一者的一些實施例中,減少表現係指相較於參考樣本、參考細胞、參考組織、對照樣本、對照細胞或對照組織,藉由標準領域已知方法(諸如本文中所描述的彼等)偵測之生物標記物(例如,蛋白或核酸(例如,基因或mRNA))之含量之約10%、20%、30%、40%、50%、60%、70%、80%、90%、95%、96%、97%、98%、99%或更大中之任一者之總體減少。在某些實施例中,減少表現係指樣本中生物標記物之表現含量/量減少,其中減少為參考樣本、參考細胞、參考組織、對照樣本、對照細胞或對照組織中相應生物標記物之表現含量/量的至少約0.9×、0.8×、0.7×、0.6×、0.5×、0.4×、0.3×、0.2×、0.1×、0.05×或0.01×中之任一者。 樣本中各種生物標記物之存在及/或表現含量/量可藉由大量方法分析,其中許多方法為此項技術中已知且熟習此項技術者瞭解,包括(但不限於)免疫組織化學(「IHC」)、西方墨點分析(Western blot analysis)、免疫沈澱、分子結合分析、ELISA、ELIFA、螢光活化細胞分選(「FACS」)、MassARRAY、蛋白質組研究、基於血液之定量分析(如例如血清ELISA)、生物化學酶活性分析、原位雜交、南方分析(Southern analysis)、北方分析(Northern analysis)、全基因組測序、聚合酶鏈反應(「PCR」)(包括定量即時PCR (「qRT-PCR」)及其他擴增類型偵測方法,諸如分支鏈DNA、SISBA、TMA及其類似物)、RNA-Seq、FISH、微陣列分析、基因表現概況分析及/或基因表現之連續分析(「SAGE」)以及可藉由蛋白質、基因及/或組織陣列分析進行的各種分析中之任一者。用於評估基因及基因產物之狀態的典型方案發現於例如Ausubel等人編., 1995, Current Protocols In Molecular Biology, Units 2 (Northern Blotting), 4 (Southern Blotting), 15 (Immunoblotting)and 18(PCR Analysis)中。亦可使用多工免疫分析,諸如可自Rules Based Medicine或Meso Scale Discovery (「MSD」)獲得之免疫分析。 在一些實施例中,生物標記物之存在及/或表現含量/量使用包含以下之方法確定:(a)對樣本(諸如受試者癌症樣本)進行基因表現概況分析、PCR (諸如rtPCR)、RNA-seq、微陣列分析、SAGE、MassARRAY技術或FISH;以及b)確定樣本中生物標記物之存在及/或表現含量/量。在一些實施例中,微陣列方法包含使用微陣列晶片,該微陣列晶片具有可在嚴格條件下與編碼上述基因之核酸分子雜交的一或多種核酸分子,或具有可結合於由上述基因編碼之一或多種蛋白質的一或多種多肽(諸如肽或抗體)。在一個實施例中,PCR方法為qRT-PCR。在一個實施例中,PCR方法為多工PCR。在一些實施例中,基因表現藉由微陣列量測。在一些實施例中,基因表現藉由qRT-PCR量測。在一些實施例中,基因表現藉由多工PCR量測。 用於評估細胞中之mRNA的方法為所熟知的且包括例如使用互補DNA探針之雜交分析(諸如使用對一或多種基因具有特異性之經標記核糖核酸探針之原位雜交、北方墨點及相關技術)及各種核酸擴增分析(諸如使用對基因中之一或多者具有特異性之互補引子之RT-PCR,及其他擴增類型偵測方法,諸如分支鏈DNA、SISBA、TMA及其類似物)。 來自哺乳動物之樣本可適宜使用北方、點漬墨法或PCR分析來對mRNA進行分析。此外,此等方法可包括允許測定生物樣本中之靶標mRNA之含量的一或多個步驟(例如,藉由同時檢查「管家」基因(諸如肌動蛋白家族成員)之比較性對照mRNA序列的含量)。視情況,可測定經擴增靶標cDNA之序列。 任選的方法包括藉由微陣列技術檢查或偵測組織或細胞樣本中之mRNA(諸如靶標mRNA)之方案。使用核酸微陣列,來自測試及對照組織樣本之測試及對照mRNA樣本經反轉錄及標記以產生cDNA探針。探針隨後經與固定於固體支撐物上之核酸陣列雜交。陣列經組態使得陣列之各成員之序列及位置為已知的。舉例而言,其表現與增加或減少之抗血管生成療法之臨床效益相關之一系列基因可排列於固體支撐物上。經標記探針與特定陣列成員之雜交指示衍生探針之樣本表達彼基因。 根據一些實施例,藉由觀測前述基因之蛋白表現含量來量測存在及/或表現含量/量。在某些實施例中,該方法包含使生物樣本在允許結合生物標記物之條件下與本文所描述之生物標記物之抗體接觸,且偵測在抗體與生物標記物之間是否形成複合物。此方法可為活體外或活體內方法。在一個實施例中,抗體用於選擇適合於具有BTK抑制劑之療法的受試者,例如用於選擇個體的生物標記物。 在某些實施例中,使用IHC及染色方案來偵測樣本中生物標記物蛋白質之存在及/或表現含量/量。組織切片之IHC染色經展示為測定或偵測樣本中之蛋白質之存在的可靠方法。在方法、分析法及/或套組中之任一者的一些實施例中,漿母細胞生物標記物選自IgJ、Mzb1及Txndc5中之一或多者。在一些實施例中,藉由免疫組織化學偵測到IgJ、Mzb1及/或Txndc5。在一些實施例中,來自個體之樣本中之漿母細胞生物標記物之高表現為高蛋白表現,且在其他實施例中係使用IHC測定。在一個實施例中,使用包含以下步驟之方法來測定生物標記物之表現含量:(a)對具有抗體之樣本執行IHC分析;以及b)測定樣本中之生物標記物之表現含量。在一些實施例中,相對於參考來測定IHC染色強度。在一些實施例中,參考為參考值。在一些實施例中,參考為參考樣本(例如,對照細胞株染色樣本)。在一些實施例中,組織為腎組織。在其他實施例中,使用螢光原位雜交代替IHC來執行上文技術。 IHC可與其他技術(諸如形態染色及/或螢光原位雜交)組合執行。兩種IHC方法為可用的;直接分析及間接分析。根據第一分析,直接地測定抗體至靶標抗原的結合。此直接分析使用經標記反應劑,諸如螢光標記或經酶標記之初級抗體,其可經目測而不需其他抗體相互作用。在典型的間接分析中,未結合初級抗體結合至抗原且隨後經標記二級抗體結合至初級抗體。當二級抗體結合至酶標記時,添加顯色或螢光受質以提供抗原之目測。由於若干二級抗體可與初級抗體上之不同抗原決定基反應,因此出現信號放大。 用於IHC之初級及/或二級抗體通常將由可偵測部分標記。若干標記為可用,其可大體上分組為以下類別:(a)放射性同位素,諸如35 S、14 C、125 I、3 H及131 I;(b)膠狀金粒子;(c)螢光標記,包括(但不限於)稀土螯合物(銪螯合物)、德克薩斯紅(Texas Red)、若丹明(rhodamine)、螢光素(fluorescein)、丹醯基(dansyl)、麗絲胺(Lissamine)、傘酮(umbelliferone)、藻紅蛋白(phycocrytherin)、藻藍蛋白(phycocyanin)或可商購之螢光團,諸如SPECTRUM ORANGE7及SPECTRUM GREEN7及/或上述中之任何一或多者之衍生物;(d)各種酶受質標記為可用的簽且美國專利第4,275,149號提供此等中之一些之評述。酶標記之實例包括螢光素酶(例如,螢火蟲螢光素酶及細菌螢光素酶;美國專利第4,737,456號)、螢光素、2,3-二氫酞嗪二酮、蘋果酸鹽去氫酶、尿素酶、過氧化酶(諸如辣根過氧化酶(horseradish peroxidase;HRPO))、鹼性磷酸酶、β-半乳糖、澱粉酶、溶菌酶、糖氧化酶(例如,葡萄糖氧化酶、半乳糖氧化酶及葡萄糖-6-磷酸去氫酶)、雜環氧化酶(諸如尿酸酶及黃嘌呤氧化酶)、乳過氧化酶、微過氧化酶及其類似物。 酶受質組合之實例包括例如具有作為受質之氫過氧化酶的辣根過氧化酶(HRPO);具有作為顯色受質之磷酸對硝苯酯的鹼性磷酸酶(AP);以及具有顯色受質(例如,對硝苯基-β-D-半乳糖)或螢光受質(例如,4-甲基傘形基-β-D-半乳糖)之β-D-半乳糖(β-D-Gal)。對於此等之一般評述,參看美國專利第4,275,149號及第4,318,980號。 在方法中之任一者的一些實施例中,藉由免疫組織化學使用診斷性抗體(亦即,初級抗體)偵測到漿母細胞生物標記物。在一些實施例中,待分析組織為腎組織。在一些實施例中,診斷性抗體特異性結合IgJ、Mzb1或Txndc5。在診斷性抗體中之任一者的一些實施例中,診斷性抗體為非人類抗體。在一些實施例中,診斷性抗體為大鼠、小鼠或家兔抗體。在一些實施例中,診斷性抗體為單株抗體。在一些實施例中,診斷性抗體經直接地標記。 在替代性方法中,可使樣本在足以形成抗體生物標記物複合物之條件下與對該生物標記物具有特異性之抗體接觸,且隨後偵測該複合物。可以多種方式,諸如藉由西方墨點法及用於分析廣泛多種組織及樣本(包括血漿或血清)之ELISA程序來偵測生物標記物之存在。使用此類分析型式之廣泛範圍之免疫分析技術為可用的,參看例如美國專利第4,016,043號、第4,424,279號及第4,018,653號。此等分析包括非競爭類型之單點及兩點或「夾心(sandwich)」分析,以及傳統競爭性結合分析。此等分析亦包括經標記抗體與標靶生物標記物之直接結合。 亦可藉助於功能性或活性類分析來偵測組織或細胞樣本中經選擇生物標記物之存在及/或表現含量/量。舉例而言,若生物標記物為酶,則吾人可進行此項技術中已知之分析以測定或偵測組織或細胞樣本中給定酶活性之存在。 在某些實施例中,針對所分析生物標記物之量及所使用樣本之質量之可變性之差異及運行之分析之間的可變性兩者來標準化樣本。此標準化可藉由偵測且併入某些標準化生物標記物,包括熟知之管家基因(諸如ACTB)之表現來實現。可替代地,標準化可基於所有經分析基因或其較大子集之平均或中間信號(全面標準化方式)。在逐基因基礎(gene-by-gene basis)上,將所量測的個體樣本mRNA或蛋白質之標準化量與參考集中所發現之量進行比較。用於各mRNA或蛋白/測試樣本/受試者之標準化表現含量可表現為參考集中所量測之表現含量百分比。待分析之特定受試者樣本中所量測之存在及/或表現含量/量將在此範圍內降低一些百分點,其可藉由此項技術中熟知之方法測定。 在某些實施例中,如下測定基因之相對表現含量: 相對表現基因1樣本1=2 exp (Ct管家基因-Ct基因1)以及樣本中測定之Ct。 相對表現基因1參考RNA=2 exp (Ct管家基因-Ct基因1)以及參考樣本中測定之Ct。 標準化相對表現基因1樣本1=(相對表現基因1樣本1/相對表現基因1參考RNA)×100 Ct為臨限值循環。Ct為循環數,在其處,反應內產生之螢光超出臨限值線。 所有實驗標準化為參考RNA,其為來自各種組織來源(例如,來自Clontech,Mountain View,CA之參考RNA #636538)之RNA的全面混合。相同參考RNA包括於各qRT-PCR運行中,從而允許比較不同實驗運行之間的結果。 在一個實施例中,樣本為臨床樣本。在另一實施例中,樣本用於診斷性分析。在一些實施例中,樣本獲自組織。組織活檢體通常用於獲得代表性組織碎片。可替代地,可間接地獲得呈組織或液體形式之腫瘤細胞,已知或認為該等組織或液體含有所關注細胞。可自組織或自其他身體樣本(諸如尿、痰、血清或血漿)偵測到基因或基因產物。藉由篩檢此等身體樣本,可更容易藉由測試用於靶標基因或基因產物之此等身體樣本來監測療法之進展。 在某些實施例中,參考樣本、參考細胞、參考組織、對照樣本、對照細胞或對照組織為單個樣本或來自相同受試者或個體之組合的多個樣本,在除獲得測試樣本外之一或多個不同時間點處獲得該多個樣本。舉例而言,參考樣本、參考細胞、參考組織、對照樣本、對照細胞或對照組織在比獲得測試樣本更早的時間點處自相同受試者或個體獲得。若在疾病之初始診斷期間獲得參考樣本且當疾病發展時隨後獲得測試樣本,則此參考樣本、參考細胞、參考組織、對照樣本、對照細胞或對照組織可為有用的。 在某些實施例中,參考樣本、參考細胞、參考組織、對照樣本、對照細胞或對照組織為來自不為受試者或個體之一或多個健康個體的經合併之多個樣本。在某些實施例中,參考樣本、參考細胞、參考組織、對照樣本、對照細胞或對照組織為來自不為受試者或個體之患有疾病或病症之一或多個個體的經合併之多個樣本。在某些實施例中,參考樣本、參考細胞、參考組織、對照樣本、對照細胞或對照組織為來自不為受試者或個體之來自正常組織之經合併RNA樣本或來自一或多個個體之經合併血漿或血清樣本。在某些實施例中,參考樣本、參考細胞、參考組織、對照樣本、對照細胞或對照組織為來自不為受試者或個體之來自組織之經合併RNA樣本或來自患有疾病或病症之一或多個個體之經合併血漿或血清樣本。 在某些實施例中,參考樣本、參考細胞、參考組織、對照樣本、對照細胞或對照組織為樣本細胞株。在某些實施例中,參考樣本、參考細胞、參考組織、對照樣本、對照細胞或對照組織為血液。 在一些實施例中,樣本為來自個體之組織樣本。在一些實施例中,組織樣本為血液或尿液樣本。在一些實施例中,組織樣本為血液樣本。 在方法中之任一者的一些實施例中,BTK抑制劑為小分子BTK抑制劑。在一些實施例中,小分子BTK抑制劑為化合物(A)或其醫藥學上可接受之鹽。 在任一方法之一些實施例中,根據上文實施例中之任一者的個體或患者可為人類。 在另一實施例中,本文提供治療SLE的方法。在一個實施例中,該方法包括向患有SLE之個體投與有效量之小分子BTK抑制劑。在一個此類實施例中,該方法進一步包括向個體投與有效量之至少一種額外治療劑,如下文所描述。在一些實施例中,個體可為人類。 本文中所描述之BTK抑制劑可在療法中單獨使用或與其他試劑組合使用。例如,額外治療劑可為抗炎劑、免疫調節劑、化學治療劑、細胞凋亡增強劑、神經營養因子、治療心血管疾病之藥劑、治療肝病之藥劑、抗病毒劑、治療血液病症之藥劑、治療糖尿病之藥劑及治療免疫缺陷病症之藥劑。第二治療劑可為NSAID抗炎劑。第二治療劑可為化學治療劑。醫藥組合調配物或給藥方案之第二化合物較佳具有化合物(I)之補充活性,使得其等彼此不會有不利之影響。 在一些實施例中,額外治療劑選自由以下組成之群:皮質類固醇(例如,潑尼松(prednisone)、潑尼龍(prednisolone)、甲基潑尼龍(methylprednisolone)及氫皮質酮(hydrocortisone));疾病修飾抗風濕藥物(「DMARD」,例如免疫抑制或抗炎劑);抗瘧疾劑(例如,羥基氯奎及氯奎);免疫抑制劑(例如,環磷醯胺、硫唑嘌呤、黴酚酸嗎啉乙酯(mycophenolate mofetil)、甲胺喋呤);抗炎劑(例如,阿司匹靈(aspirin)、NSAID (例如,布洛芬(ibuprofen)、萘普生(naproxen)、吲哚美辛(indomethacin)、萘丁美酮(nabumetone)、塞內昔布(celecoxib)));抗高血壓劑(例如,鈣通道阻斷劑(例如,胺氯地平(amlodipine)、硝苯地平(nifedipine))及利尿劑(例如,呋喃苯胺酸(furosemide)));士他汀類(statin )(例如,阿托伐他汀(atorvastatin)、氟伐他汀(fluvastatin)、洛伐他汀(lovastatin)、匹伐他汀(pitavastatin)、普伐他汀(pravastatin)、羅素他汀(rosuvastatin)及辛伐他汀(simvastatin));抗B細胞劑(例如,抗CD20 (例如利妥昔單抗)、抗CD22);抗B淋巴球刺激劑(「抗BLyS」,例如貝利單抗(belimumab)、布里莫德(blisibimod));1型干擾素受體拮抗劑(例如,阿尼佛單抗(anifrolumab) );T細胞調節劑(例如,力者莫德(rigerimod));阿巴西普(abatacept);抗凝血劑(例如,肝素、華法林(warfarin));以及維生素D補充劑。 組合療法可以同時或以依序方案投與。當依序投與時,該組合可依兩次或更多次投與形式給藥。組合投與包括使用分開調配物或單一醫藥調配物共同投與,及依任何次序連續投與,其中兩種(或所有)活性劑最好在一段時間同時發揮其生物活性。以上共同投與藥劑中之任一者的適合劑量為當前所使用之劑量且可由於額外治療劑之組合作用(協同作用)而降低。 組合療法可為複合的,使得當一起使用活性成分時獲得的效應大於由單獨使用化合物產生之效應的總和。可在活性成分經以下操作時獲得協同效應:(1)同時投與或遞送;(2)交替或同時投與;或(3)藉由一些其他方案。當以交替療法遞送時,可在連續投與或遞送化合物時獲得協同效應。大體而言,在交替療法期間,依序(亦即,連續)投與有效劑量之各活性成分,然而在組合療法中,一起投與有效劑量之兩種或更多種活性成分。 在組合療法中,套組可包含(a)具有本發明之劑型組合物的第一容器及視情況,(b)具有含於其中之第二醫藥調配物以用於與本發明之劑型組合物共同投與的第二容器。在此等態樣中,套組可包含用於含有單獨的組合物之容器,諸如分割瓶或分割箔包,然而,單獨的組合物亦可包含於單一無隔膜容器內。通常,套組包含用於投與單獨的組分之指示。當單獨的組分較佳以不同劑型(例如,經口及非經腸)投與時,以不同給藥時間間隔投與時或處方醫師需要滴定組合之個別組分時,套組形式為尤其有利的。 BTK抑制劑可藉由任何適合方式投與,包括經口、非經腸、肺內及鼻內,且視需要用於局部治療之病灶內投與。在較佳實施例中,經口投與BTK抑制劑。 包含BTK抑制劑之經口劑型包括(但不限於)包含BTK抑制劑或其醫藥學上可接受之鹽及一或多種醫藥學上可接受之賦形劑的錠劑或膠囊。在一些實施例中,包含BTK抑制劑之錠劑或膠囊可根據本文所提供之方法每天投與一次或兩次。在本文所提供之某些實施例中,口服劑型為包含化合物(A)或其醫藥學上可接受之鹽及一或多種醫藥學上可接受之賦形劑的錠劑。 非經腸輸注包括肌肉內、靜脈內、動脈內、腹膜內或皮下投與。部分視投藥之短期或長期性而定,可藉由任何適合途徑(例如,藉由注射,諸如靜脈內或皮下注射)給藥。本文中涵蓋各種給藥時程,包括(但不限於)單次投藥或在不同時間點的多次投藥、快速投藥及脈衝式輸注。 本文中所描述之BTK抑制劑可以與良好醫學實踐一致的方式調配、給藥及投與。在此情形下,考慮因素包括所治療之特定疾病或病症、所治療之特定哺乳動物、個別患者之臨床病狀、疾病或病症之起因、藥劑之遞送位點、投與方法、投與時程及醫學從業者已知之其他因素。BTK抑制劑無需但視情況與一或多種當前用於預防或治療疾病或病症之藥劑一起調配。此等其他藥劑之有效量視存在於調配物中之BTK抑制劑之量、疾病或病症或治療之類型及如上文所論述之其他因素而定。此等藥劑通常以相同劑量且以如本文所描述之投與途徑使用,或以本文所描述之劑量之約1%至99%使用,或以任何劑量且藉由經驗/臨床上測定為合適的任何途徑使用。 III. 包含 BTK 抑制劑之 治療性組合物 本文中之組合物、方法及套組提供小分子BTK抑制劑。本文所提供之小分子BTK抑制劑較佳地為除結合多肽或抗體外之有機分子,且可使用已知方法鑑別及化學合成。結合有機小分子通常大小小於約2000道爾頓,可替代地大小小於約1500、750、500、250或200道爾頓,其中能夠結合(較佳地特定言之)如本文所描述之BTK之此等有機小分子可使用熟知技術鑑別且不需過度實驗。就此而言,應指出用於篩檢用於可結合於多肽靶標之分子之有機小分子庫的技術為此項技術中所熟知的(參看例如PCT公開案第WO 2000/00823號及第WO 2000/39585號)。結合有機小分子可為例如醛、酮、肟、腙、半卡腙、卡肼、一級胺、二級胺、三級胺、N取代之肼、醯肼、醇、醚、硫醇、硫醚、二硫化物、羧酸、酯、醯胺、脲、胺基甲酸酯、碳酸酯、縮酮、硫縮酮、縮醛、硫縮醛、芳基鹵化物、磺酸芳酯、烷基鹵化物、磺酸烷酯、芳族化合物、雜環化合物、苯胺、烯烴、炔烴、二醇、胺基醇類、噁唑啶、噁唑啉、噻唑啶、噻唑啉、烯胺、磺醯胺、環氧化物、氮丙啶、異氰酸酯、磺醯基氯化物、重氮化合物、酸氯化物或其類似者。 在方法中之任一者的一些實施例中,BTK抑制劑選自由以下組成之群:依魯替尼(ibrutinib)、阿卡拉布魯替尼(acalabrutinib)、斯比布魯替尼(spebrutinib)、BIIB068 (Biogen)、BMS-986195 (Bristol-Myers Squibb)、BMS-986142 (Bristol-Myers Squibb)、BMS-935177 (Bristol-Myers Squibb)、M2951 (Merck KGaA)、PRN-1008 (Principia Biopharma)、HM71224/LY3337641 (Hanmi/Lilly)、ONO-4059/GS-4059 (Gilead/Ono)、AC0058 (ACEA Biosciences)、AC0025 (ACEA Biosciences)、ABBV-599 (AbbVie)、ABBV-105 (AbbVie)、PF-303 (Pfizer)、BI-BTK1 (Boehringer Ingelheim)、CC90008 (Celgene)、AS550 (Carna Biosciences)、ARQ 531 (Arqule)、AEG42766 (Aegera Therapeutics)、BGB-3111 (Beigene)、RN486 (Simcere Pharma)、HCI-1401 (LSK BioPharma/Hustman Cancer Inst.)、KBP-7536 (KBP Bioscience)、RDX002 (RedX Biopharma)、SNS-062 (Sunesis)、TAS5315 (Taiho Pharma)、TAX-020 (Takeda)、WX486/WXFL-10230486 (WuXi AppTec/Humanwell)及X-022 (X-Rx Discovery)。 在一些實施例中,BTK抑制劑為化合物(A)或其醫藥學上可接受之鹽。本文提供之BTK抑制劑之醫藥學上可接受之鹽可用於本文之方法中。如本文中所使用,術語「醫藥學上可接受之鹽」意欲包括用相對無毒之酸或鹼製備之活性化合物的鹽,其視本文所描述之化合物上所存在之特定取代基而定。當本發明化合物含有相對酸性官能基時,可藉由使此類化合物之中性形式在無溶劑下或在適合的惰性溶劑中與足夠量之所需鹼接觸來獲得鹼加成鹽。衍生自醫藥學上可接受之無機鹼之鹽的實例包括鋁鹽、銨鹽、鈣鹽、銅鹽、鐵鹽、亞鐵鹽、鋰鹽、鎂鹽、錳鹽、亞錳鹽、鉀鹽、鈉鹽、鋅鹽及其類似鹽。衍生自醫藥學上可接受之有機鹼的鹽包括一級、二級及三級胺之鹽,包括經取代之胺、環狀胺、天然存在之胺及其類似物,諸如精胺酸、甜菜鹼、咖啡因、膽鹼、N,N'-二苯甲基乙二胺、二乙胺、2-二乙胺基乙醇、2-二甲胺基乙醇、乙醇胺、乙二胺、N-乙基嗎啉、N-乙基哌啶、還原葡糖胺、葡糖胺、組胺酸、海卓胺(hydrabamine)、異丙胺、離胺酸、甲基還原葡糖胺、嗎啉、哌嗪、哌啶、多元胺樹脂、普魯卡因(procaine)、嘌呤、可可豆鹼、三乙胺、三甲胺、三丙胺、緩血酸胺及其類似物。當本發明之化合物含有相對鹼性官能基時,可藉由使此等化合物之中性形式在無溶劑下或在適合惰性溶劑中與足夠量之所要酸接觸來獲得酸加成鹽。醫藥學上可接受之酸加成鹽的實例包括衍生自無機酸之彼等酸加成鹽,該等無機酸如鹽酸、氫溴酸、硝酸、碳酸、一氫碳酸、磷酸、一氫磷酸、二氫磷酸、硫酸、一氫硫酸、氫碘酸或亞磷酸及類似酸;以及衍生自相對無毒之有機酸之鹽,該等有機酸如乙酸、丙酸、異丁酸、丙二酸、苯甲酸、丁二酸、辛二酸、反丁烯二酸、扁桃酸、鄰苯二甲酸、苯磺酸、對甲苯基磺酸、檸檬酸、酒石酸、甲磺酸及類似者。亦包括諸如精胺酸及類似酸之胺基酸的鹽,及如葡糖醛酸或半乳糖醛酸及其類似酸之有機酸的鹽(參見例如Berge, S. M.等人, 「Pharmaceutical Salts」, Journal of Pharmaceutical Science, 1977, 66, 1-19)。本發明之某些特定化合物含有允許化合物轉化成鹼加成鹽或酸加成鹽之鹼性及酸性官能基兩者。 化合物之中性形式可藉由使鹽與鹼或酸接觸且以習知方式分離親本化合物來再生。化合物之親本形式與各種鹽形式的不同之處在於某些物理特性,諸如在極性溶劑中之溶解性,但出於本發明之目的,在其他方面,鹽等效於化合物之親本形式。 除鹽形式以外,本發明提供呈前藥形式之化合物。如本文中所使用,術語「前藥」係指容易在生理學條件下發生化學改變以提供本發明之化合物的彼等化合物。另外,前藥可藉由化學方法或生物化學方法在離體環境中轉化成本發明之化合物。舉例而言,當前藥與適合的酶或化學試劑一起置於經皮貼片儲集層中時,其可緩慢轉化成本發明之化合物。 本發明之前藥包括化合物,其中胺基酸殘基或兩種或更多種(例如兩種、三種或四種)胺基酸殘基之多肽鏈經由醯胺或酯鍵共價接合至本發明之化合物之游離胺基、羥基或羧酸基團。胺基酸殘基包括(但不限於) 20種通常由三個字母符號表示的天然存在之胺基酸,且亦包括磷酸絲胺酸、磷酸蘇胺酸、磷酸酪胺酸、4-羥基脯胺酸、羥基離胺酸、鎖鏈離胺酸、異鎖鏈離胺酸、γ-羧基麩胺酸、馬尿酸、八羥吲哚-2-羧酸、他汀(statine)、1,2,3,4-四氫異喹啉-3-羧酸、青黴胺、鳥胺酸、3-甲基組胺酸、正纈胺酸、β-丙胺酸、γ-胺基丁酸、瓜胺酸、高半胱胺酸、高絲胺酸、甲基-丙胺酸、對苯甲醯基苯基丙胺酸、苯基甘胺酸、炔丙基甘胺酸、肌胺酸、甲硫胺酸碸及第三丁基甘胺酸。 亦涵蓋其他類型之前藥。舉例而言,本發明化合物之游離羧基可衍生為醯胺或烷基酯。作為另一實例,包含游離羥基之本發明化合物可藉由將羥基轉化成諸如(但不限於)磷酸酯、半丁二酸酯、二甲基胺基乙酸酯或磷醯基氧甲基氧羰基之基團而衍生為前藥,如Fleisher, D.等人,(1996) Improved oral drug delivery: solubility limitations overcome by the use of prodrugs Advanced Drug Delivery Reviews, 19:115中所概述。亦包括羥基及胺基之胺基甲酸酯前藥,如同碳酸酯前藥,亦包括羥基之磺酸酯及硫酸酯。亦涵蓋衍生為(醯氧基)甲基及(醯氧基)乙基醚之羥基,其中醯基可為視情況經包括(但不限於)醚、胺及羧酸官能基之基團取代的烷基酯,或其中醯基為如上文所描述之胺基酸酯。此類型之前藥描述於J. Med. Chem ., (1996), 39:10中。更特定實例包括用以下基團置換醇基之氫原子:諸如(C1 -6 )烷醯氧基甲基、1-((C1 -6 )烷醯氧基)乙基、1-甲基-1-((C1 -6 )烷醯氧基)乙基、(C1 -6 )烷氧基羰氧基甲基、N-(C1 -6 )烷氧基羰基胺基甲基、丁二醯基、(C1 -6 )烷醯基、α-胺基(C1 -4 )烷醯基、芳醯基及α-胺基醯基或α-胺基醯基-α-胺基醯基,其中各α-胺基醯基獨立地選自天然存在之L-胺基酸、P(O)(OH)2 、-P(O)(O(C1 -6 )烷基)2 或糖基(由移除碳水化合物之半縮醛形式之羥基產生的基團)。 對於前藥衍生物之額外實例,參見例如a) Design of Prodrugs,由H.Bundgaard編輯,(Elsevier, 1985)及Methods in Enzymology, 第42卷. 第309-396頁, 由K.Widder等人編輯(Academic Press, 1985);b) A Textbook of Drug Design and Development,由Krogsgaard-Larsen及H. Bundgaard編輯, 第5章「Design and Application of Prodrugs」, 由H. Bundgaard編輯, 第113-191頁(1991)頁;c) H. Bundgaard, Advanced Drug Delivery Reviews, 8:1-38 (1992);d) H. Bundgaard, 等人, Journal of Pharmaceutical Sciences, 77:285 (1988);及e) N. Kakeya等人, Chem. Pharm. Bull., 32:692 (1984),其中各者特定地以引用之方式併入本文中。 本發明之某些化合物可以未溶劑化形式以及溶劑化形式(包括水合形式)存在。大體而言,溶劑化形式等效於未溶劑化形式,且意欲包涵於本發明範疇內。某些本發明化合物可以多種結晶形式或非晶形式存在。大體而言,所有物理形式皆等效地用於本發明涵蓋之用途且意欲在本發明範疇內。 本發明之某些化合物具有不對稱碳原子(光學中心)或雙鍵;外消旋體、非對映異構體、幾何異構體、區位異構體及個別異構體(例如,單獨的對映異構體)皆意欲涵蓋於本發明之範疇內。IV. 醫藥調配物 BTK抑制劑之醫藥調配物提供於本文之方法及套組中。 在方法之中之任一者的一些實施例中,基於患者體重以約0.1毫克/公斤/天至約100毫克/公斤/天、約0.5毫克/公斤/天至約20毫克/公斤/天,或約1毫克/公斤/天至約10毫克/公斤/天之劑量投與BTK抑制劑(例如,化合物(A)或其醫藥學上可接受之鹽)。在一些實施例中,以約10至800 mg之劑量投與呈錠劑之化合物(A)或其醫藥學上可接受之鹽。在一些實施例中,以約25至300 mg之劑量投與呈錠劑中之游離鹼之化合物(A)。在一些實施例中,錠劑包含25至300 mg之呈游離鹼之化合物(A)及反丁烯二酸,其中化合物(A)與反丁烯二酸之重量比為約1:5至約3:1;或約1:2至約2:1;或約1:1.5至約1.5:1。在一些實施例中,錠劑包含25至300 mg之呈游離鹼之化合物(A)及反丁烯二酸,且其中反丁烯二酸含量為約5重量%至約50重量%、約5重量%至約40重量%、約5重量%至約30重量%、約10重量%至約30重量%、約20重量%至約25重量%、約5重量%至約15重量%或約10重量%至約15重量%。在以上實施例中之一些中,錠劑重量為約100 mg、約200 mg、約300 mg、約400 mg、約500 mg、約600 mg、約700 mg、約800 mg、約900 mg或約1000 mg。在一些實施例中,錠劑進一步包含選自填充劑、結合劑、崩解劑、潤滑劑及滑動劑之至少一種醫藥學上可接受之賦形劑。在一些實施例中,錠劑包含乳糖及微晶纖維素。 本發明之錠劑組合物可進一步適當地包含選自(但不限於)填充劑(稀釋劑)、崩解劑、結合劑、滑動劑及潤滑劑之一或多種醫藥學上可接受之賦形劑。填充劑(或稀釋劑)可用於增加構成錠劑之粉末藥物的總體積。崩解劑可用於在攝入錠劑時促進錠劑分解為較小碎片,理想地為個體藥物粒子,且由此有助於藥物之快速溶解及吸收。結合劑可用於確保顆粒及錠劑可形成具有所需機械強度且在已經壓縮之後使錠劑保存在一起,預防其在包裝、送貨及常規操作期間分解為其組分粉末。滑動劑可用於在生產期間改良構成錠劑之粉末的流動性。潤滑劑可用於確保在製造期間製錠粉末不會黏附於用於按壓錠劑之設備,以在混合及按壓期間改良粉末之流動,且在最終錠劑自設備噴出時最小化摩擦及破裂。 填充劑及結合劑可包括磷酸氫鈣、微晶纖維素(Avicel®)、乳糖或任何其他適合膨化劑。適合填充劑之實例包括微晶纖維素,諸如Avicel PH 101、Avicel PH102、Avicel PH 200、Avicel PH 105、Avicel DG、Ceolus KG 802、Ceolus KG 1000、SMCCSO及Vivapur 200;單水合乳糖,諸如乳糖FastFlo;與其他賦形劑共加工之微晶纖維素,諸如與乳糖單一水合物(MicroceLac 100)共加工之微晶纖維素及與膠態二氧化矽(SMCCSO、Prosolv 50及Prosolv HD 90)共加工之微晶纖維素;異麥芽酮糖衍生物之混合物,諸如galenIQ;以及其他適合填充劑及其組合。填充劑可以顆粒內組分及/或顆粒外組分之形式存在。在一些特定態樣中,本發明之錠劑組合物包含乳糖及微晶纖維素。 崩解劑可包括於所揭示之調配物中以促進壓製品內之顆粒彼此分離且維持所釋放顆粒彼此分離。崩解劑可以顆粒內組分及/或顆粒外組分之形式存在。崩解劑可包括任何適合崩解劑諸如交聯聚合物,諸如交聯聚乙烯吡咯啶酮及交聯羧基甲基纖維素鈉或交聯羧甲纖維素鈉。在一些特定態樣中,崩解劑為交聯羧甲纖維素鈉。崩解劑含量適當地為約1重量%、約1.5重量%、約2重量%、約2.5重量%、約3重量%、約3.5重量%、約4重量%、約4.5重量%、或約5重量%及其範圍,諸如約1重量%至約5重量%或約2重量%至約4重量%。 滑動劑可包括例如膠態二氧化矽,包括高度分散之氧化矽(Aerosil®)或任何其他適合滑動劑,諸如動物或植物脂肪或蠟。在一些特定態樣中,滑動劑為煙霧狀氧化矽。滑動劑含量適當地為約0.1重量%、約0.5重量%、約1重量%、約1.5重量%、約2重量%、約2.5重量%或約3重量%,及其範圍,諸如約0.1重量%至約3重量%、約0.5重量%至約2重量%、約0.5重量%至約1.5重量%。 潤滑劑可用於壓緊醫藥組合物中之顆粒。潤滑劑可包括例如聚乙二醇(例如,分子量為約1000至約6000)、硬脂酸鎂及硬脂酸鈣、硬脂醯反丁烯二酸鈉、滑石或任何其他適合潤滑劑。在一些特定態樣中,潤滑劑為硬脂酸鎂及/或硬脂醯反丁烯二酸鈉。潤滑劑可以顆粒內組分及/或顆粒外組分之形式存在。潤滑劑含量適當地為約0.5重量%、約1重量%、約1.5重量%、約2重量%、約2.5重量%、約3重量%、約3.5重量%、約4重量%、約4.5重量%或約5重量%及其範圍,諸如約0.5重量%至約5重量%、約1重量%至約4重量%、約1重量%至約3重量%或約1重量%至約2重量%。 可將包衣(諸如膜包衣)塗覆至本發明之錠劑。膜包衣可用於例如幫助錠劑可易於吞咽。亦可採用膜包衣以改良口味及外觀。若需要,膜包衣可為腸衣。膜包衣可包含聚合性成膜材料,諸如羥丙基甲基纖維素、羥丙基纖維素、丙烯酸酯或甲基丙烯酸脂共聚物及聚乙烯醇-聚乙二醇接枝共聚物,諸如Opadry及Kollicoat IR。除成膜聚合物以外,膜包衣可另外包含塑化劑,例如聚乙二醇;界面活性劑,例如Tween®型;及視情況選用之顏料,例如二氧化鈦或氧化鐵。膜包衣亦可包含滑石作為抗黏附劑。膜包衣典型地佔小於約5重量%之劑型。 本文之調配物亦可含有超過一種為所治療之特定適應症所必需之活性成分,較佳為具有不會對彼此產生不利影響之補充性活性的彼等活性成分。此類活性成分宜以有效達成預期目的之量的組合存在。 可例如藉由凝聚技術或藉由界面聚合將活性成分包覆於所製備之微囊中,例如羥基甲基纖維素或明膠微囊及聚(甲基丙烯酸甲酯)微囊分別包覆於膠態藥物遞送系統(例如,脂質體、白蛋白微球體、微乳液、奈米粒子及奈米膠囊)或巨乳液中。此等技術揭示於Remington's Pharmaceutical Sciences 第16版, Osol, A.編(1980)中。 可製備延釋製劑。延釋製劑之適合實例包括含有BTK抑制劑之固體疏水性聚合物之半滲透基質,該等基質呈成形物品之形式,例如膜或微囊。 用於活體內投與之調配物通常為無菌的。無菌性可容易地藉由例如用無菌過濾膜過濾來實現。V. 製品 在另一實施例中,提供一種含有適用於治療、預防及/或診斷上文所述之病症之材料的製品。製品包含容器及容器上或與容器相關聯之標記或藥品說明書。適合容器包括例如瓶子、小瓶、注射器、IV溶液袋等。容器可由各種材料形成,諸如玻璃或塑膠。容器容納某一組合物本身或某一組合物與可有效治療、預防及/或診斷病狀之另一組合物之組合,且可具有無菌接取口(例如容器可為具有可由皮下注射針刺穿之塞子的靜脈內溶液袋或小瓶)。組合物中之至少一種活性劑為本文中所描述的BTK抑制劑。標記或藥品說明書指示組合物用於治療所選病狀。此外,製品可包含(a)第一容器以及其中所含之組合物,其中該組合物包含BTK抑制劑;以及(b)第二容器以及其中所含之組合物,其中該組合物包含另一細胞毒性或另外的治療劑。 在一些實施例中,製品包含容器、該容器上之標記及包含於該容器內之組合物;其中該組合物包括一或多種反應劑(結合至一或多種生物標記物或本文中所描述的生物標記物中之一或多者之探針及/或引子的初級抗體(例如,B-9 Santa Cruz Biotechnology抗體)),容器上之標記指示組合物可用以評估樣本中一或多種生物標記物之存在,及用於使用反應劑以供評估樣本中一或多種生物標記物之存在的指令。製品可進一步包含用於製備樣本及利用反應劑的一組指令及材料。在一些實施例中,製品可包括反應劑,諸如初級抗體及二級抗體兩者,其中二級抗體結合至標記,例如酶標記。在一些實施例中,針對本文中所描述的生物標記物中之一或多者之製品、一或多種探針及/或引子。 此實施例中之製品可進一步包含指示組合物可使用以治療特定病狀之藥品說明書。替代地或另外,製品可進一步包含第二(或第三)容器,其包含醫藥學上可接受之緩衝液,諸如抑菌性注射用水(BWFI)、磷酸鹽緩衝鹽水、林格氏溶液(Ringer's solution)及右旋糖溶液。其可進一步包括就商業及使用者觀點而言所需之其他材料,包括其他緩衝劑、稀釋劑、過濾器、針及注射器。 製品中之其他視情況選用之組分包括一或多種緩衝液(例如,阻斷緩衝液、洗滌緩衝液、受質緩衝液等)、其他反應劑(諸如藉由酶標記化學上改變之受質) (例如色原體)、抗原決定基恢復溶液、對照樣本(正及/或負對照)、對照切片等。實例 以下為方法及組合物之實例。應瞭解,考慮到上文提供之一般描述,可實施各種其他實施例。血液樣本分析 3- 基因漿母細胞標籤表現分析。 將血液收集於PAXgene RNA試管(PreAnalytiX)中;使用可商購套組根據製造商說明書(Qiagen)提取總RNA。生物標記物: IgJ、TXNDC5、MZB1。參考基因: TMEM55B 藉由Human Genome U133 Plus 2.0陣列(Affymetrix Inc., Santa Clara, CA)評估血液樣本中之候選生物標記基因之表現。由Asuragen Inc.(Austin,TX)執行微陣列雜交。原始CEL檔案資料係使用Robust Multi-array Averaging (RMA)求和及標準化且使用R及Bioconductor進行分析。 可替代地,藉由Fluidigm qPCR分析來定量血液樣本中之候選生物標記基因之表現。3種基因得分係根據IgJ、TXNDC5及MZB1之平均值計算,且使用參考基因TMEM55B標準化。使用Cobas 4800平台(Roche Molecular Systems)上產生之此分析來評估血液樣本。實例 1 漿母細胞轉錄組之特徵 對活體外經分化之CD20loCD38+漿母細胞、CD20+CD27+活化B細胞及CD20+CD27-原生B細胞執行轉錄分析以鑑別在B細胞子集之間具有較強分化表現之基因(圖1A-1)。在0.001之假髮現率(FDR)下,鑑別出86種基因,該等基因在漿母細胞中之表現比活化B細胞或原生B細胞中之任一者高>10倍。執行此等資料之進一步改進以僅包括在漿母細胞中具有>5 nRPKM之基因,得到總共40種基因。許多此等基因包括重鏈及輕鏈區段,以及涉及免疫球蛋白蛋白質之生物合成的基因。選擇不為免疫球蛋白基因座之部分的生物標記物候選物,因此自候選基因清單移除此等候選物(圖1B)。 經證實,藉由執行活體外漿母細胞分化分析由布魯東氏酪胺酸激酶(BTK)活性來調節漿母細胞分化,其中將人類記憶B細胞經置於分化條件下,且隨後使用流式細胞量測術定量在5天之後量測CD20loCD38++漿母細胞。以劑量依賴性方式使用特定且強力BTK激酶活性抑制劑(GDC-0852)來抑制CD40L誘導之漿母細胞分化(圖7)。GDC-0852為(S)-2-(5-氟基-2-(羥甲基)-3-(1-甲基-5-(5-(2-甲基-4-(氧雜環丁-3-基)哌嗪-1-基)吡啶-2-基胺基)-6-側氧基-1,6-二氫吡啶-3-基)-苯基)-3,4,6,7,8,9六氫吡啶并[3,4-b]吲哚嗪-1(2H)-酮,其結構如下展示:。 為確保所選生物標記物候選物可準確地測定具有高敏感性及特異性之樣本中之漿母細胞之分率,對衍生自3個供體之活體外區分之漿母細胞之40,000至39個細胞執行連續2倍稀釋成1,000,000個衍生自兩個單獨供體之PBMC。使用Fluidigm評估候選漿母細胞標記基因之表現含量,報導為相對於管家基因HPRT1之ΔCt。線性回歸用於建模由log10漿母細胞頻率預測之候選基因之ΔCt,其中漿母細胞供體及PBMC供體作為共變量。大部分吾等候選基因展示與漿母細胞頻率之較強關聯,及漿母細胞供體之間的最小差異。三種基因,IGJ、MZB1及TXNDC5表現特別良好,分別地展示0.84、0.75及0.69之r2 (圖2B-1、2B-2、2B-3、2B-4)。取三種基因之平均值作為標籤分數產生r2 為0.79之標籤分數。 為驗證活體內分化漿母細胞中之基因標籤,量測其在直接地自五個一週接種流感痘苗之健康供體分選之漿母細胞中之表現。全部三種候選生物標記基因相對於原生及記憶B細胞兩者在漿母細胞中更高度表現(圖2C-1、2C-2、2C-3、2C-4)。實例2:漿母細胞標記基因與活體內漿母細胞頻率相關 在來自ROSE II期臨床試驗[3]之狼瘡患者群中使用流式細胞量測術來量測全血中之漿母細胞之頻率,其具有成對RNA定序資料。所選漿母細胞標籤基因經展示顯示與IgD-CD19+ CD27++ CD38++ 漿母細胞之頻率之高相關性(圖2D-1、2D-2、2D-3、2D-4)。IGJ、MZB1及TXNDC5展示與漿母細胞含量之最高相關係數,斯皮爾曼相關性係數分別為0.66、0.71及0.71。取三種標籤基因之平均值作為標籤展示與漿母細胞頻率之較強相關性(斯皮爾曼ρ=0.71)。此等結果表明可使用所選三種基因標籤在全血樣本中量測漿母細胞之相對豐度。 實例3:漿母細胞標籤與SLE之增強的疾病活動性相關 先前工作已表明漿母細胞頻率與疾病嚴重程度之間的較強相關性,該疾病嚴重程度如由紅斑狼瘡中雌激素安全性(SELENA)-全身性紅斑性狼瘡症疾病活動指數(SLEDAI)得分所量測。此相關性似乎由患有靜態疾病及活動性疾病之患者中之差異驅動。觀測來自EXPLORER II期臨床試驗[4]之中度至重度腎外狼瘡群,發現漿母細胞標籤展示與疾病活動性之較低但顯著相關性(斯皮爾曼ρ=0.19,p=0.03,圖3A)。更加仔細地觀測驅動此相關性之SLEDAI組分,發現特定言之三種子得分與高漿母細胞豐度尤其相關聯:DNA結合、低補體及淋巴球減少症(圖3B-1、圖3B-2、圖3B-3)。 在多個狼瘡患者群中之補體組分C3及C4之漿母細胞豐度與血清濃度之間發現中度負相關性(斯皮爾曼ρ分別=-0.34、-0.38,圖3C)。亦觀測到此等群中之漿母細胞含量與抗雙股DNA抗體之效價之間的中度相關性(斯皮爾曼ρ=0.39,圖3C-1、圖3C-2、圖3C-3)。 大部分狼瘡患者展示干擾素活性之轉錄標籤[2,5]。漿母細胞標籤展示與使用三種基因標籤來量測之干擾素活性的中度相關性(圖3D-1及3D-2;[5])。相關性似乎由具有高含量之干擾素活性之患者子集中之升高的漿母細胞標籤表現驅動,其中大部分較低干擾素標籤患者具有較低漿母細胞基因表現,而具有高干擾素活性之患者展示較低含量漿母細胞基因表現及高含量漿母細胞基因表現之混合。然而,漿母細胞標籤獨立於干擾素標籤而與疾病嚴重程度及血清學活性相關;使用後向模型選擇,其中Akaike資訊準則作為度量,漿母細胞及干擾素標籤預測血清補體含量且漿母細胞標籤單獨預測抗dsDNA抗體效價及SLEDAI。 無論作為組合得分或針對個體疾病領域,未在漿母細胞標籤分數與不列顛群島狼瘡評估小組(British Isles Lupus Assessment Group;BILAG)活性指數之間看到關聯。此等資料支持漿母細胞在由自體抗體、淋巴球減少症及低補體血症(hypocomplementemia)驅動之血清學疾病活動性中之作用。實例4:利妥昔單抗治療減少漿母細胞標籤 自來自在SLE中評估利妥昔單抗之安全性及效能的II期臨床試驗的兩個中度至重度狼瘡患者群中收集漿母細胞標籤值。該等群為患有狼瘡性腎炎(LUNAR)或腎外狼瘡(EXPLORER)之患者[4,6]。在治療療程內混合效應建模漿母細胞標籤值,併入共變量年齡、人種、合併用藥、干擾素活性、SLEDAI、問診、治療隊組及其相互作用,其中患者建模為隨機效應,鑑別出在經利妥昔單抗治療之患者內具有特異性之漿母細胞標籤的大幅減少(圖4A、圖4B)。此效應在接受利妥昔單抗輸注兩週後之時間點中最顯而易見,且隨時間推移效應消減。在EXPLORER試驗中,在第四次輸注利妥昔單抗兩週後,在第28週觀測到3.48倍之最大減少(p=2×10-11 )。同樣,在LUNAR試驗中,在28週時間點處觀測到最低含量之漿母細胞標籤表現, 具有3.31倍減少(p=0.0011)。 在EXPLORER試驗中,監測患者之針對小鼠人類嵌合抗體(HACA)之抗體的存在。雖然大部分利妥昔單抗治療之患者展示漿母細胞標籤之減少,觀測到繼續出現抗藥物抗體之患者展示在利妥昔單抗治療之後受限於漿母細胞標籤之無減少圖4C)。如上所述,使用相同線形混合效應模型,在繼續出現HACA之患者中發現2.8倍高之漿母細胞標記基因表現(p=0.0007)。實例5:狼瘡標準護理治療改變漿母細胞標籤 先前研究已鑑別出不同免疫抑制治療與減少之漿母細胞相關基因表現之間的關聯[7]。觀測來自EXPLORER臨床試驗之篩檢樣本,相較於經硫唑嘌呤治療之患者,在經黴酚酸酯或甲胺喋呤治療之患者中發現極低之漿母細胞標籤表現(圖5A)。在基線處進行黴酚酸酯治療之患者中觀測到來自LUNAR試驗之患者中漿母細胞標籤基因表現降低的趨勢(圖5B),儘管相對少患者在MMF治療集區中。實例6:患者人種/種族影響漿母細胞標籤含量 生物標記物含量在患者群體之間常常不同。資料分析顯示在EXPLORER及ROSE臨床試驗群體兩者中相對於非洲或美籍西班牙人血統患者之歐洲血統之患者中之顯著較低之漿母細胞標籤含量。當考慮干擾素活性、年齡及疾病嚴重程度時,此為正確的(圖6A、圖6B、圖6C)。實例7:BTK抑制對漿母細胞分化之效應 漿母細胞分化:自健康供體PBMC (Miltenyi記憶B細胞分離套組)分離記憶B細胞。對於漿母細胞分化,隨後在細胞介素、IL-2 (20 U/ml)、IL-10 (50 ng/ml)、IL-15 (10 ng/ml)、IL-6 (50 ng/ml)、IFNa (10 ng/ml)之混合物之存在下,培育1.5×10^5/ml記憶B細胞,且用ODN2006 (TLR-9配位體) 5 ug/ml或CD40L (3 ug/ml)刺激5天。在單獨媒劑(DMSO)及呈各種濃度之GDC-0852的存在下進行漿母細胞分化,使用在10 uM開始的3倍劑量之抑制劑滴定。執行流式細胞量測術以枚舉(CD20- CD38++ )漿母細胞百分比且評估GDC-0852之抑制。 RNA製備:自在5天時的含細胞之培養物提取RNA,該含細胞之培養物係來自DMSO及GDC-0852(370 nM)處理之細胞(n=4)。使用Qiashredder (Qiagen, Valencia, CA)在RLT緩衝液中破壞細胞,且隨後使用包括柱上DNase消化之RNeasy微型套組(Qiagen)提取RNA。使用NanoDrop 8000 (Thermo Scientific)測定總RNA之濃度及RNA樣本之完整性。經分離RNA係用於Fluidigm定量RT-PCR分析。 QT PCR:使用iScript cDNA合成套組(Biorad, Hercules, CA)對100 ng總-RNA執行cDNA合成。對3種基因(IgJ、MZB1、TXNDC5,包括管家基因TMEM55B)執行基因特異性預擴增(Applied Biosystems)。使用BioMark 48.48動態陣列(Fluidigm Corporation)使用製造商之方案執行RT-PCR。使用生物標記資料收集軟體(BioMark Data Collection Software)收集資料且使用生物標記RT-PCR分析軟體(BioMark RT-PCR Analysis Software) (V.2.1.1,Fluidigm)獲得CT值。計算HPRT1之相對豐度(dCt):2log-(平均Ct基因-平均Ct HPRT1)。對於統計分析,將低於偵測下限之值設置為比最低記錄值低1 Ct。 使用以R程式化語言編寫之<>或自定義腳本執行統計分析。為鑑別基因表現中之差異,吾等使線形混合效應模型擬合至log2轉化之相對轉錄豐度,其中治療作為固定效應且供體作為隨機效應。為比較DMSO與化合物處理樣本之間的漿母細胞百分比,吾等使用威爾科克森等級求和測試(Wilcoxon rank sum test)。使用格拉夫帕德稜鏡軟體(GraphPad Prism software)計算BTK抑制之IC50值。人類重組介白素(IL)-2及干擾素-α (IFN-α)購自R&D系統(Minneapolis, MN)且IL-10、IL-6及IL-15購自Peprotech (Rocky Hill, NJ)。CpG (ODN2006)購自Invivogen (San Deigo, CA)且CD40L購自R&D系統(Minneapolis, MN)。結果 將B細胞分化成漿母細胞可經由多個活化刺激發生且涉及不同分子變化。經由CD40及/或類鐸受體(Toll like TLR)之活化B細胞使得將CD20+ CD27++ 記憶B細胞分化為CD20- CD38++ 漿母細胞。吾等藉由使用CD40L刺激之T細胞介導之反應或使用類鐸受體配位體CpG之T細胞獨立反應來評估BTK抑制劑GDC-0852在漿母細胞分化方法中之效應。 GDC-0852以劑量依賴方式在第5天時抑制CD40L誘導之漿母細胞分化,其中IC50效能為20.0nM (+/-0.002) (圖7)。經DMSO及GDC-0852處理之細胞的基因表現分析展示漿母細胞標籤基因IgJ(p 0.011 )、MZB1 (p 0.0023 )、TXNDC5 (p 0.0032 )及複合漿母細胞3種基因標籤(p 0.0026 )之顯著減少,同時原生B細胞展示極低之標籤基因表現含量(p<1×10-6 ) (圖8)。將基因表現值與漿母細胞豐度進行比較,吾等發現3種基因標籤與漿母細胞百分比之間的較強相關性(斯皮爾曼ρ=0.81) (圖9)。CpG介導之漿母細胞分化(n=3)亦由GDC-0852抑制,其中IC50效能為48 nM (+/-57) (圖10)。 當引入本發明或其較佳實施例之要素時,冠詞「一(a,an)」、「該(the)」及「該(said)」意謂存在該等要素中之一或多者。術語「包含(comprising)」、「包括(including)」及「具有(having)」意欲為包容性的,且意謂可能存在除所列要素之外的其他要素。 儘管出於清楚理解之目的,已藉助於說明及實例相當詳細地描述前述本發明,但該等描述及實例不應解釋為限制該範疇。本文引用的所有專利及科學文獻之揭示內容以全文引用之方式明確地併入本文中。 參考文獻 1. Arce E, Jackson DG, Gill MA, Bennett LB, Banchereau J, Pascual V. Increased frequency of pre-germinal center B cells and plasma cell precursors in the blood of children with systemic lupus erythematosus. J Immunol. 2001;167: 2361-2369. doi:10.4049/jimmunol.167.4.2361 2. Bennett L, Palucka a K, Arce E, Cantrell V, Borvak J, Banchereau J, et al. Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med. 2003;197: 711-723. doi:10.1084/jem.20021553 3. Kalunian KC, Merrill JT, Maciuca R, McBride JM, Townsend MJ, Wei X, et al. A Phase II study of the efficacy and safety of rontalizumab (rhuMAb interferon-α) in patients with systemic lupus erythematosus (ROSE). Ann Rheum Dis. 2016;75: 196-202. doi:10.1136/annrheumdis-2014-206090 4. Merrill JT, Neuwelt CM, Wallace DJ, Shanahan JC, Latinis KM, Oates JC, et al. Efficacy and safety of rituximab in moderately-to-severely active systemic lupus erythematosus: The randomized, double-blind, phase II/III systemic lupus erythematosus evaluation of rituximab trial. Arthritis Rheum. 2010;62: 222-233. doi:10.1002/art.27233 5. Kennedy WP, Maciuca R, Wolslegel K, Tew W, Abbas AR, Chaivorapol C, et al. Association of the interferon signature metric with serological disease manifestations but not global activity scores in multiple cohorts of patients with SLE. Lupus Sci Med. 2015;2: e000080. doi:10.1136/lupus-2014-000080 6. Rovin BH, Furie R, Latinis K, Looney RJ, Fervenza FC, Sanchez-Guerrero J, et al. Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: the Lupus Nephritis Assessment with Rituximab study. Arthritis Rheum. 2012;64: 1215-1226. doi:10.1002/art.34359 7. Banchereau R, Hong S, Cantarel B, Baldwin N, Baisch J, Edens M, et al. Personalized Immunomonitoring Uncovers Molecular Networks that Stratify Lupus Patients. Cell. Elsevier Inc.; 2016;165: 551-565. doi:10.1016/j.cell.2016.03.008 I. definition As used interchangeably herein, "polynucleotide" or "nucleic acid" refers to a polymer of nucleotides of any length and includes DNA and RNA. Nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases and / or their analogs, or can be incorporated into polymers by DNA or RNA polymerase or by synthetic reactions Any of them. Polynucleotides may include modified nucleotides, such as methylated nucleotides and the like. If present, modifications to the nucleotide structure can be imparted before or after assembly of the polymer. The nucleotide sequence may be interspersed with non-nucleotide components. Polynucleotides can be further modified after synthesis, such as in conjunction with a label. Other types of modifications include, for example, "capping"; replacement of one or more naturally occurring nucleotides with analogs; internucleotide modifications, such as having uncharged linkages (e.g., methyl phosphonates, phosphate triesters, phosphorus Amines, carbamates, etc.) and others with charged linkages (e.g., phosphorothioate, phosphorodithioate, etc.), containing proteins such as nucleases, toxins, antibodies, signal peptides, Poly-L-lysine, etc.), flanking parts, those with intercalating agents (e.g., acridine, psoralen, etc.), chelating agents (e.g., metals, radioactive metals, boron, oxidation) Metals, etc.), those containing alkylating agents, those having modified linkages (e.g., alpha-rotomers, etc.), and unmodified forms of polynucleotides. In addition, any hydroxyl group typically present in a sugar may be replaced, for example, by a phosphonate group, a phosphate group, protected by a standard protecting group, or activated to make additional linkages with additional nucleotides, or may be bound to a solid or Semi-solid support. The 5 'and 3' terminal OH can be phosphorylated or partially substituted with an amine or organic capping group of 1 to 20 carbon atoms. Other hydroxy groups can also be derived into standard protecting groups. Polynucleotides may also contain similar forms of ribose or deoxyribose commonly known in the art, including, for example, 2'-O-methyl-, 2'-O-allyl, 2'-fluoro-, or 2 '-Azido-ribose, carbocyclic analogs, alpha-arameloses, epimers (such as arabinose, xyloses, or lyxoses); piperazine Pranose, furanose, sedoheptuloses, acyclic analogs, and abasic nucleoside analogs (such as methyl riboside). One or more phosphodiester bonds may be replaced by alternative linking groups. Such alternative linking groups include, but are not limited to, phosphoric acid via P (O) S ("thiothioate"), P (S) S ("dithioate"), (O) NR2 (`` Methylamine ''), P (O) R, P (O) OR ', CO or CH2 ("Formal") embodiment of substitution, wherein each R or R 'is independently H or optionally contains an ether (-O-) bond, aryl, alkenyl, cycloalkyl, cycloalkenyl or aromatic Alkyl substituted or unsubstituted alkyl (1-20 C). Not all bonds in a polynucleotide need to be consistent. The foregoing description applies to all polynucleotides mentioned herein, including RNA and DNA. As used herein, "oligonucleotide" generally refers to a shorter single-stranded polynucleotide, but need not be less than 250 nucleotides in length. Oligonucleotides can be synthetic. The terms "oligonucleotide" and "polynucleotide" are not mutually exclusive. The above description of polynucleotides is equally and fully applicable to oligonucleotides. The term "primer" refers to a single-stranded polynucleotide that is capable of hybridizing to a nucleic acid, and then polymerizing with a complementary nucleic acid, usually by providing a free 3'-OH group. The term "small molecule" refers to any molecule having a molecular weight of about 2000 Daltons or less, preferably about 500 Daltons or less. The terms "host cell", "host cell line", and "host cell culture" are used interchangeably and refer to a cell into which a foreign nucleic acid has been introduced, including descendants of such cells. Host cells include "transformants" and "transformed cells", which include primary transformed cells and descendants derived from them, regardless of the number of generations. Progeny may not have the same nucleic acid content as the mother cell, but may contain mutations. This article includes screening or selecting mutant offspring that have the same function or biological activity against primitively transformed cells. As used herein, the term "vector" refers to a nucleic acid molecule capable of delivering another nucleic acid to which it is linked. The term includes vectors that serve as self-replicating nucleic acid structures as well as vectors incorporated into the genome of a host cell into which they have been introduced. Certain vectors are capable of directing the performance of nucleic acids to which they are operatively linked. These vectors are referred to herein as "expression vectors". An "isolated" antibody is one that has been separated from components of its natural environment. In some embodiments, the antibody is purified to, for example, as determined by, for example, electrophoresis (e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatography (e.g., ion exchange or reverse phase HPLC). 95% or 99% purity. For a review of methods for assessing antibody purity, see, for example, Flatman et al.,J. Chromatogr. B 848: 79-87 (2007). "Isolated" nucleic acid refers to a nucleic acid molecule that has been separated from components of its natural environment. An isolated nucleic acid includes a nucleic acid molecule contained in a cell that usually contains a nucleic acid molecule, but the nucleic acid molecule is present outside the chromosome or at a chromosomal location different from its natural chromosomal location. The term "antibody" is used herein in the broadest sense and encompasses a variety of antibody structures including, but not limited to, monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments, as long as they are It is sufficient to exhibit the desired antigen-binding activity. A "blocking" or "antagonist" antibody is an antibody that inhibits or reduces the biological activity of the antigen to which it binds. It is preferred that the blocking or antagonist antibody substantially or completely inhibit the biological activity of the antigen. "Affinity" refers to the strength of the sum of non-covalent interactions between a single binding site of a molecule (eg, an antibody) and its binding partner (eg, an antigen). Unless otherwise indicated, as used herein, "binding affinity" refers to the intrinsic binding affinity that reflects a 1: 1 interaction between members of a binding pair (eg, an antibody and an antigen). The affinity of molecule X for its partner Y is generally expressed by the dissociation constant (Kd). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described below. An "affinity maturity" anti-system refers to an antibody having such changes in one or more hypervariable regions compared to a parent antibody that does not have one or more changes in one or more hypervariable regions (HVR). These changes improve the affinity of the antibody for the antigen. The term "detection" includes any detection method, including direct and indirect detection. As used herein, the term "biomarker" refers to an indicator that can be detected in a sample, such as predictive, diagnostic, and / or prognostic. Biomarkers can serve as indicators of specific subtypes that are characterized by certain molecular, pathological, histological, and / or clinically characteristic diseases or conditions (eg, cancer). In some embodiments, the biomarker is a gene. Biomarkers include, but are not limited to, polynucleotides (e.g., DNA and / or RNA), polypeptides, polypeptide and polynucleotide modifications (e.g., post-translational modifications), carbohydrate and / or glycolipid molecular markers Thing. The terms "biomarker tag", "tag", "biomarker performance tag" or "performance tag" are used interchangeably herein and refer to a biomarker or a combination thereof whose performance is indicative Properties, such as predictive, diagnostic, and / or prognostic. A biomarker tag can serve as an indicator of a particular subtype characterized by a disease or condition (eg, cancer) of certain molecular, pathological, histological, and / or clinical characteristics. In some embodiments, the biomarker tag is a "gene tag." The term "gene tag" is used interchangeably with "gene expression tag" and refers to a polynucleotide, or a combination thereof, that behaves as an indicator, such as predictive, diagnostic, and / or prognostic. In some embodiments, the biomarker tag is a "protein tag." The term "protein tag" is used interchangeably with "protein expression tag" and refers to a polypeptide, or combination thereof, that behaves as an indicator, such as predictive, diagnostic, and / or prognostic. The "amount" or "content" of a biomarker associated with an increased clinical benefit to an individual is the detectable amount in the sample. These can be measured by methods known to those skilled in the art and also disclosed herein. The performance content or amount of the assessed biomarker can be used to determine the response to treatment. The terms "expressed content" or "expressed content" are generally used interchangeably and generally refer to the amount of a biomarker in a biological sample. "Performance" generally refers to the method by which information (e.g., genetic coding and / or epigenetics) is transformed into a structure that exists in and operates in a cell. Thus, as used herein, "performance" may refer to transcription into a polynucleotide, translation into a polypeptide, or even polynucleotide and / or polypeptide modification (eg, post-translational modification of a polypeptide). Transcribed polynucleotides, translated polypeptides, or fragments of polynucleotides and / or polypeptide modifications (e.g., post-translational modifications of polypeptides) should also be considered performance, whether derived from transcripts or degradation resulting from alternative splicing The transcripts are also derived, for example, from post-translational processing of proteolytic peptides. "Expression genes" include genes that are transcribed into polynucleotides such as mRNA and subsequently translated into polypeptides, and also genes that are transcribed into RNA but not translated into polypeptides (eg, transfer and ribosomal RNA). "High performance", "High performance content" or "High content" refers to a biomarker in an individual relative to a control (such as an individual or an individual without an illness or disorder (e.g., cancer) or an internal control (e.g., housekeeper Biomarkers)) increased expression or increased content. "Reduced performance", "reduced performance content" or "reduced content" refers to a biomarker in an individual relative to a control (such as an individual or an individual without an illness or disorder (e.g., cancer) or an internal control (e.g., housekeeper Biomarkers)). In some embodiments, the reduced performance is minimal or no performance. In certain embodiments, the term "under reference content" refers to the content of a biomarker in a sample from an individual or patient that is substantially equivalent to the reference content or refers to a difference of up to 1%, up to 2% from the reference content , Up to 3%, up to 4%, up to 5%. In some embodiments, the reference content is an intermediate content of a biomarker in a reference population. In some embodiments, the reference content of the marker is the average content of the marker in the reference population. In some embodiments, the reference content of the marker is the average content of the marker in the reference population. In certain embodiments, the term "above reference content" refers to at least 5%, 10%, 20%, 25%, 30% above the reference content as determined by the methods described herein relative to the reference content. , 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95%, 100% or more of the biomarker content in a sample from an individual or patient. In some embodiments, the reference content is an intermediate content in a reference population. In some embodiments, the reference content of the marker is the average content of the marker in the reference population. In certain embodiments, the term "below the reference content" means at least 5%, 10%, 20%, 25%, 30% below the reference content, as determined by the methods described herein, relative to the reference content. , 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95%, 100% or more of the biomarker content in a sample from an individual or patient. In some embodiments, the reference content is an intermediate content in a reference population. In some embodiments, the reference content of the marker is the average content of the marker in the reference population. In some embodiments, the reference content of the marker is the average content of the marker in the reference population. The term "housekeeper biomarker" refers to a biomarker or population of biomarkers (eg, polynucleotides and / or polypeptides) that are typically similarly found in all cell types. In some embodiments, the housekeeping biomarker is a "housekeeping gene". A "housekeeping gene" refers herein to a gene or group of genes that encodes a protein that is essential for the maintenance of cell function and is typically similarly present in all cell types. As used herein, "amplification" generally refers to a method of generating multiple copies of a desired sequence. "Multiple copies" means at least two copies. "Duplicate" does not necessarily mean a perfect sequence that is complementary or consistent with the template sequence. For example, replicas may include nucleotide analogs, such as deoxyinosine, intentional sequence changes (such as sequence changes introduced via primers that include sequences that are hybridizable to the template but are not complementary), and / or occur during amplification Sequence error. The term "multiplex PCR" refers to a single PCR reaction performed on a nucleic acid obtained from a single source (eg, an individual) using more than one primer for the purpose of amplifying two or more DNA sequences in a single reaction. The "stringency" of the hybridization reaction can be easily determined by those skilled in the art, and is usually calculated empirically depending on the length of the probe, washing temperature, and salt concentration. In general, longer probes require higher temperatures for proper annealing, while shorter probes require lower temperatures. When complementary strands are present in an environment below their melting temperature, hybridization usually depends on the ability of the denatured DNA to reanneal. The higher the desired degree of homology between the probe and the hybridizable sequence, the higher the relative temperature at which it can be used. Therefore, it can be seen that the higher the relative temperature will tend to make the reaction conditions more stringent, and the temperature will also decrease. For additional details and explanations of the stringency of hybridization reactions, see Ausubel et al., Current Protocols in Molecular Biology, Wiley Interscience Publishers, (1995). As defined herein, "stringent conditions" or "high stringency conditions" can be identified by: (1) the use of low ionic strength and high temperature for washing, for example at 50 ° C, 0.015 M sodium chloride / 0.0015 M sodium citrate / 0.1% sodium lauryl sulfate; (2) using a denaturing reagent such as formamidine during hybridization, for example, at 42 ° C, with 0.1% bovine serum albumin / 0.1% ficol (Ficoll) /0.1% polyvinylpyrrolidone / 50 mM sodium phosphate buffer at pH 6.5 and 50% (v / v) formamidine of 750 mM sodium chloride and 75 mM sodium citrate; or (3) at 42 50% formamidine, 5 × SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 × Denhardt's solution at ℃ , Sonicated salmon sperm DNA (50 μg / ml), 0.1% SDS and 10% dextran sulfate in hybridization overnight, with 42 ° C in 0.2 × SSC (sodium chloride / sodium citrate) A 10 minute wash was followed by a 10 minute high stringency wash consisting of 0.1 x SSC containing EDTA at 55 ° C. "Medium stringent conditions" can be identified as described by Sambrook et al., Molecular Cloning: A Laboratory Manual, New York: Cold Spring Harbor Press, 1989, and include the use of washing solutions and more relaxed than those described above. Hybridization conditions (eg, temperature, ionic strength, and% SDS). Examples of moderately stringent conditions are incubation overnight at 37 ° C in a solution containing: 20% formamidine, 5 × SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5 × Dunhart's solution, 10% dextran sulfate, and 20 mg / ml denatured sheared salmon sperm DNA, and then the filtrate was washed in 1 × SSC at about 37-50 ° C. Those skilled in the art will know how to adjust temperature, ionic strength, etc. as necessary to accommodate factors such as probe length and the like. The term "diagnosis" is used herein to refer to the identification or classification of a molecular or pathological condition, disease, or condition (eg, cancer). For example, "diagnosis" can refer to the identification of a specific type of cancer. "Diagnosis" may also refer to a subtype characterized, for example, by histopathological criteria, or by the performance of a molecular feature (e.g., by a biomarker (e.g., a specific gene or protein encoded by the gene) or a combination thereof ) Classify cancer specific subtypes. The term "assisted diagnosis" refers herein to a method that assists in making a clinical determination regarding the presence or nature of a particular type of symptom or condition of a disease or disorder (eg, cancer). For example, a method to assist in the diagnosis of a disease or condition (e.g., cancer) may include measuring certain biomarkers in a biological sample from an individual. As used herein, the term "sample" refers to a composition obtained or derived from a subject and / or individual of interest, which contains, for example, discriminable and / or physical, biochemical, chemical and / or physiological characteristics Cells and / or other molecular entities identified. For example, the phrase "disease sample" and its variants refers to any sample obtained from a subject of interest that is expected or known to contain cells and / or molecular entities with characteristics to be identified. Samples include (but are not limited to) native or cultured cells or cell lines, cell supernatants, cell lysates, platelets, serum, plasma, vitreous fluid, lymph fluid, synovial fluid, follicular fluid, semen, amniotic fluid , Milk, whole blood, blood-derived cells, urine, cerebrospinal fluid, saliva, sputum, tears, sweat, mucus, tumor lysates and tissue culture media, tissue extracts (such as homogenized tissue), tumor tissue, Cell extracts and combinations thereof. "Tissue sample" or "cell sample" means a collection of similar cells obtained from the tissue of a subject or individual. The source of the tissue or cell sample may be solid tissue, such as: from fresh, frozen and / or preserved organs, tissue samples, biopsies and / or extracts; blood or any blood component such as plasma; body fluids such as cerebrospinal fluid , Amniotic fluid, peritoneal fluid, or interstitial fluid; cells from a subject at any time during pregnancy or development. Tissue samples can also be native or cultured cells or cell lines. Optionally, a tissue or cell sample is obtained from the diseased tissue / organ. Tissue samples may contain compounds such as preservatives, anticoagulants, buffers, fixatives, nutrients, antibiotics, or the like, which do not naturally intermix with tissues of nature. As used herein, "reference sample", "reference cell", "reference tissue", "control sample", "control cell" or "control tissue" means a sample, cell, tissue, standard used for comparison purposes Or content. In one embodiment, the reference sample, reference cell, reference tissue, control sample, control cell, or control tissue is obtained from a healthy and / or disease-free portion (e.g., tissue or cell) of the same subject or individual's body . For example, healthy and / or non-diseased cells or tissues near diseased cells or tissues (eg, cells or tissues near tumors). In another embodiment, the reference sample is obtained from untreated tissue and / or cells of the same subject or individual's body. In yet another embodiment, the reference sample, reference cell, reference tissue, control sample, control cell, or control tissue is obtained from a healthy and / or non-diseased part of the body of another individual who is not the subject or individual ( (E.g., tissue or cell). In yet another embodiment, the reference sample, reference cell, reference tissue, control sample, control cell, or control tissue is obtained from untreated tissue and / or cells of the body of another individual who is not the subject or individual. For the purposes of this document, a "slice" of a tissue sample means a single portion or fragment of a tissue sample, such as a thin layer of tissue or cells cut from a tissue sample. It should be understood that multiple sections of a tissue sample can be used and analyzed, with the limitation being that it should be understood that the same section of a tissue sample can be analyzed in both morphology and molecular content, or in terms of both polypeptides and polynucleotides analysis. "Correlate / correlating" means comparing the performance and / or results of the first analysis or protocol with the performance and / or results of the second analysis or protocol in any way. For example, we can use the results of the first analysis or protocol for the second protocol and / or we can use the results of the first analysis or protocol to determine whether the second analysis or protocol should be performed. Regarding examples of polynucleotide analysis or protocol, we can use the results of the polynucleotide performance analysis or protocol to determine whether a particular treatment protocol should be performed. "Individual response" or "response" can be assessed using any endpoint that indicates an individual benefit, including (but not limited to): (1) inhibiting the progression of disease (e.g., cancer progression) to a certain extent, including slowing Complete suppression; (2) Reduction of tumor size; (3) Inhibition (i.e., reduction, slowing, or completion of suppression) of cancer cells penetrating close to surrounding organs and / or tissues; (4) Inhibition (i.e., reduction, reduction (Slow or complete suppression) lesions; (5) relieve to some extent one or more symptoms associated with a disease or disorder (e.g., cancer); (6) increase the length of progression-free survival; and / or (7) during treatment The given time point is followed to reduce mortality. As used herein, the term "substantially the same" means a sufficiently high degree of similarity between two values, so that in the case of biological characteristics measured by such values (e.g., Kd value or performance), familiarity with this The skilled person will consider the difference between the two values to be minimal or non-biological and / or statistically significant. The difference between the two values is, for example, less than about 50%, less than about 40%, less than about 30%, less than about 20%, and / or less than about 10%, which varies with reference / comparative values. As used herein, the phrase "substantially different" indicates a sufficiently high degree of difference between the two values, so that in the case of biological characteristics measured by such values (e.g., Kd value), familiarize yourself with this The skilled person will consider the difference between these two values to be statistically significant. The difference between the two values is, for example, greater than about 10%, greater than about 20%, greater than about 30%, greater than about 40%, and / or greater than about 50%, which varies with the value of the reference / comparison molecule. The phrase "label" as used herein means a detectable compound or composition. The label is typically bound or fused directly or indirectly to a reagent, such as a polynucleotide probe or antibody, and helps to detect the reagent to which it is bound or fused. The label itself can be detectable (e.g., a radioisotope label or a fluorescent label), or in the case of an enzyme label, it can catalyze a chemical change in the host compound or composition to produce a detectable product. An "effective amount" of an agent is an amount effective to achieve the desired therapeutic or preventive result at the required dose and time period. The "therapeutically effective amount" of a substance / molecule (agonist or antagonist) can vary depending on factors such as the disease condition, age, sex, and weight of the individual, and the substance / molecule (agonist or antagonist) in the individual The ability to trigger the desired reaction. A therapeutically effective amount is also one in which any therapeutically beneficial effect exceeds any toxic or deleterious effect of the substance / molecule (agonist or antagonist). A "prophylactically effective amount" means an amount effective to achieve a desired preventive result at the required dose and time period. Since individuals use prophylactic doses before or early in the disease, a prophylactically effective amount is usually (but not necessarily) smaller than a therapeutically effective amount. The term "pharmaceutical formulation" refers to a formulation in a form that permits the biological activity of the active ingredients contained therein to be effective and which does not contain other components that have unacceptable toxicity to the individual to whom the formulation is to be administered. "Pharmaceutically acceptable carrier" means an ingredient in a pharmaceutical formulation that is not toxic to an individual other than the active ingredient. Pharmaceutically acceptable carriers include, but are not limited to, buffers, excipients, stabilizers, or preservatives. As used herein, "treatment" (and its grammatical variations, such as "treat / treating") refers to a clinical intervention that attempts to alter the natural course of an individual being treated, and may be for prevention or control purposes or Performed during the course of clinical pathology. The required therapeutic effects include, but are not limited to, preventing the occurrence or recurrence of a disease, alleviating symptoms, alleviating any direct or indirect pathological consequences of the disease, preventing cancer metastasis, slowing the rate of disease progression, improving or alleviating the disease condition, and alleviating or improving the prognosis. In some embodiments, the antibodies are used to delay or slow disease progression. As used in this application, the term "prodrug" refers to a precursor or derivative of a pharmaceutically active substance that is less cytotoxic to tumor cells than the parent drug and is capable of being activated enzymatically or Conversion to a more active parent form. See, e.g., Wilman, "Prodrugs in Cancer Chemotherapy"Biochemical Society Transactions , 14, pp. 375-382, 615th Meeting Belfast (1986) and Stella et al., "Prodrugs: A Chemical Approach to Targeted Drug Delivery",Directed Drug Delivery Borchardt et al., Eds., Pp. 247-267, Humana Press (1985). Prodrugs of the present invention include (but are not limited to) phosphate-containing prodrugs, thiophosphate-containing prodrugs, sulfate-containing prodrugs, peptide-containing prodrugs, D-amino acid modified prodrugs, and glycosylated prodrugs , Β-lactamamine-containing prodrugs, optionally substituted phenoxyacetamide prodrugs, or optionally substituted phenylacetamide prodrugs, 5-fluorocytosine and can be converted into more active Other 5-fluorouridine prodrugs of cytotoxic free drugs. Examples of cytotoxic drugs that can be derived into prodrug forms for use in the present invention include, but are not limited to, their chemotherapeutic agents described above. "Individual" or "subject" is a mammal. Mammals include, but are not limited to, domestic animals (e.g., cattle, sheep, cats, dogs, and horses), primates (e.g., human and non-human primates, such as monkeys), rabbits, and rodents (e.g., , Mouse and rat). In certain embodiments, the individual or subject is human. The term "simultaneously" is used herein to refer to the administration of two or more therapeutic agents, wherein at least a portion of the administrations overlap in time. Thus, simultaneous administration includes a dosing regimen when administration of one or more agents is continued after interruption of administration of one or more other agents. "Reducing or inhibiting" means the ability to reduce overall by 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95% or more. Reduction or inhibition can refer to the symptoms of the condition being treated, the presence or size of cancer metastases, or the size of the primary tumor. The term "pharmaceutical instructions" is used to refer to the instructions normally included in the commercial packaging of therapeutic products, which contain indications, usage, dosage, administration, combination therapy, contraindications and / Or warnings. "Article of manufacture" is any manufactured article (e.g., a package or container) or kit containing at least one reactant, such as a drug used to treat a disease or disorder (e.g., cancer) or used to specifically detect as described herein Biomarker probe. In some embodiments, the article of manufacture or kit is marketed, distributed, or sold in the form of a unit for performing the methods described herein. The phrase "based on" as used in this article means that information about one or more biomarkers is used to inform treatment decisions and information provided on the drug label or sales / promotion guide. As understood by those skilled in the art, references to "about" a value or parameter herein include (and describe) embodiments directed to that value or parameter itself. For example, a reference to "about X" includes a description of "X" itself. It should be understood that the aspects and embodiments described herein include "composition" and / or "substantially composed of" aspects and embodiments. Unless otherwise specified, as used herein, the singular forms "a / an" and "the" include plural references. II. Method and use Provided herein are methods for utilizing plasmablast biomarkers. In particular, methods using BTK inhibitors and plasmablast biomarkers. For example, methods are provided for treating an individual with a disease or condition, the methods comprising administering to the individual effective treatment if the individual has been found to have the presence and / or increased content of a plasmablast biomarker Amount of BTK inhibitor. In addition, provided herein is a method for treating a disease or condition in an individual, the method comprising: determining that a sample from the individual contains an elevated content of plasmablast biomarkers, and administering to the individual an effective amount of a BTK inhibitor, by This treats a disease or condition. In some embodiments, the plasmablast biomarker is selected from the group consisting of the following genetic tags: IgJ, Mzb1, and Txndc5. In some embodiments, the genes of IgJ, Mzb1 and Txndc5 are expressed as polypeptides, which are determined by measuring the mRNA content of the gene in a patient's blood relative to a reference content. In some embodiments, the disease or disorder is an autoimmune or inflammatory disease or disorder. In some embodiments, the disease or disorder is SLE. In some embodiments, the disease or condition is lupus nephritis. In some embodiments, the disease or condition is extrarenal lupus. Provided herein are methods of treating a disease or condition in an individual, the methods comprising administering to the individual an effective amount of a BTK inhibitor, wherein the treatment is based on the presence and / or increased content of a plasmablast biomarker in a sample from the individual . In some embodiments, the plasmablast biomarker is a manifestation of one or more of IgJ, Mzb1, and Txndc5. In some embodiments, the genes of IgJ, Mzb1 and Txndc5 are expressed as polypeptides, which are determined by measuring the mRNA content of the gene in a patient's blood relative to a reference content. In some embodiments, the disease or disorder is an autoimmune or inflammatory disease or disorder. In some embodiments, the disease or disorder is SLE. In some embodiments, the disease or condition is lupus nephritis. In some embodiments, the disease or condition is extrarenal lupus. Furthermore, provided herein are methods for selecting a therapy for an individual suffering from a disease or disorder, the methods comprising determining the presence and / or content of a plasmablast biomarker and based on the presence and / or content of the biomarker And choose drugs. In some embodiments, the drug is selected based on elevated levels of plasmablast biomarkers. In some embodiments, the plasmablast biomarker is selected from the group consisting of the following genetic tags: IgJ, Mzb1, and Txndc5. In some embodiments, the genes of IgJ, Mzb1 and Txndc5 are expressed as polypeptides, which are determined by measuring the mRNA content of the gene in a patient's blood relative to a reference content. In some embodiments, the disease or disorder is an autoimmune or inflammatory disease or disorder. In some embodiments, the disease or disorder is SLE. In some embodiments, the disease or condition is lupus nephritis. In some embodiments, the disease or condition is extrarenal lupus. Provided herein are methods for identifying individuals with a disease or disorder that are more or less likely to exhibit benefit from treatment comprising a BTK inhibitor, the method comprising: determining the presence of plasmablast biomarkers in a sample from the individual and / Or content where the presence and / or elevated content of plasmablast biomarkers in a sample indicates that the individual is more likely to exhibit benefit from treatment comprising a BTK inhibitor, or the absence and / or reduction of plasmablast biomarkers The amount indicates that the individual is unlikely to exhibit benefit from a treatment comprising a BTK inhibitor. In some embodiments, the plasmablast biomarker is selected from the group consisting of the following genetic tags: IgJ, Mzb1, and Txndc5. In some embodiments, the genes of IgJ, Mzb1 and Txndc5 are expressed as polypeptides, which are determined by measuring the mRNA content of the gene in a patient's blood relative to a reference content. In some embodiments, the disease or disorder is an autoimmune or inflammatory disease or disorder. In some embodiments, the disease or disorder is SLE. In some embodiments, the disease or condition is lupus nephritis. In some embodiments, the disease or condition is extrarenal lupus. Also provided herein is an assay for identifying an individual with a disease or disorder receiving a BTK inhibitor, the method comprising: (a) determining the presence and / or content of a plasmablast biomarker in a sample from the individual; ( b) BTK inhibitors are recommended based on the presence and / or content of plasmablast biomarkers. In some embodiments, BTK inhibitors are recommended based on elevated levels of plasmablast biomarkers. In some embodiments, the plasmablast biomarker is selected from the group consisting of the following genetic tags: IgJ, Mzb1, and Txndc5. In some embodiments, the genes of IgJ, Mzb1 and Txndc5 are expressed as polypeptides, which are determined by measuring the mRNA content of the gene in a patient's blood relative to a reference content. In some embodiments, the disease or disorder is an autoimmune or inflammatory disease or disorder. In some embodiments, the disease or disorder is SLE. In some embodiments, the disease or condition is lupus nephritis. In some embodiments, the disease or condition is extrarenal lupus. Provided herein is a diagnostic kit comprising one or more reagents for determining the content of a plasmablast biomarker in a sample from an individual having a disease or condition, wherein the presence of the plasmablast biomarker is detected And / or elevated levels mean improved efficacy when treating individuals with BTK inhibitors, and where a lower or substantially undetectable level of plasmablast biomarkers was detected meant that patients with BTK inhibitors were treated The effect is reduced in individuals with the disease. Articles are also provided herein that include a pharmaceutical composition containing a BTK inhibitor and a package insert packaged together, the package insert indicating that the BTK inhibitor is used to treat a disease or condition based on the performance of plasmablast biomarkers Patient. In some embodiments, the plasmablast biomarker is selected from the group consisting of the following genetic tags: IgJ, Mzb1, and Txndc5. In some embodiments, the genes of IgJ, Mzb1 and Txndc5 are expressed as polypeptides, which are determined by measuring the mRNA content of the gene in a patient's blood relative to a reference content. In some embodiments, the disease or disorder is an autoimmune or inflammatory disease or disorder. In some embodiments, the disease or disorder is SLE. In some embodiments, the disease or condition is lupus nephritis. In some embodiments, the disease or condition is extrarenal lupus. In addition, provided herein are methods for treating a disease or condition in an individual, the methods comprising administering to the individual an effective amount of a BTK inhibitor, and evaluating one or more plasma mothers in a sample from the individual during treatment with the BTK inhibitor The content of a cell biomarker (eg, compared to a reference). Methods of treating a disease or condition in an individual are also provided, the methods comprising administering to the individual an effective amount of a BTK inhibitor, wherein the treatment is based on the content of one or more plasmablast biomarkers in the sample from the individual (e.g., Compared to reference). Provided is a method for monitoring a response in a subject treated with a BTK inhibitor, the method comprising determining the content of one or more plasmablast biomarkers in a sample from the individual, wherein the reduced content of one or more plasmablasts A cell biomarker (e.g., compared to a reference) indicates that an individual is more likely to respond to a treatment comprising a BTK inhibitor with one or more plasmablast biomarkers that increase the content and / or are substantially the same as the pre-treatment content An object (eg, compared to a reference) indicates that the individual is unlikely to respond to a treatment comprising a BTK inhibitor. In some embodiments, the plasmablast biomarker is selected from the group consisting of the following genetic tags: IgJ, Mzb1, and Txndc5. In some embodiments, the genes of IgJ, Mzb1 and Txndc5 are expressed as polypeptides, which are determined by measuring the mRNA content of the gene in a patient's blood relative to a reference content. In some embodiments, the disease or disorder is an autoimmune or inflammatory disease or disorder. In some embodiments, the disease or disorder is SLE. In some embodiments, the disease or condition is lupus nephritis. In some embodiments, the disease or condition is extrarenal lupus. Also provided is a method of determining whether an individual with a disease or condition should continue or discontinue treatment with a BTK inhibitor, the method comprising measuring the content of one or more plasmablast biomarkers in a sample from the individual, which increases Determination of one or more plasmablast biomarkers (e.g., compared to a reference) at levels and / or levels substantially the same as before treatment. Individuals should discontinue treatment with a BTK inhibitor and reduce the level of one or more plasma. Blast cell biomarkers (eg, compared to a reference) determine that an individual should continue treatment with a BTK inhibitor. In some embodiments, the plasmablast biomarker is selected from the group consisting of the following genetic tags: IgJ, Mzb1, and Txndc5. In some embodiments, the genes of IgJ, Mzb1 and Txndc5 are expressed as polypeptides, which are determined by measuring the mRNA content of the gene in a patient's blood relative to a reference content. In some embodiments, the disease or disorder is an autoimmune or inflammatory disease or disorder. In some embodiments, the disease or disorder is SLE. In some embodiments, the disease or condition is lupus nephritis. In some embodiments, the disease or condition is extrarenal lupus. In some embodiments, the method comprises: (a) measuring the RNA content of one, two, or three biomarkers selected from the group consisting of IgJ, TXNDC5, and MZB1 in a biological sample from a patient; (b) adding (a) The RNA content measured in the comparison with the reference content; and (c) when the RNA content measured in (a) is higher than the reference content, the patient is identified as more likely to benefit from BTK inhibitor therapy. In some embodiments, the RNA is mRNA. In some embodiments, measuring mRNA content comprises amplification. In some embodiments, measuring mRNA content comprises quantitative PCR. In some embodiments, measuring the mRNA content includes amplifying the mRNA and detecting the amplified product, thereby measuring the mRNA content. In some embodiments, the reference content is the intermediate content of the corresponding marker in the reference population. In some embodiments, the reference content of the marker is an intermediate content of the marker in the reference population. In any of the embodiments described herein, the reference content may be the average content of the corresponding marker in the reference population. In some embodiments, the reference content of the marker is the average content of the marker in the reference population. Non-limiting exemplary reference populations include patients with immune or inflammatory diseases, healthy individuals, and populations including healthy individuals and patients with immune or inflammatory diseases. In some embodiments, the reference population comprises patients with SLE. In some embodiments, the method of analyzing or detecting a biomarker has a p-value of less than 0.05. In some embodiments, the method has a specificity greater than 80%. In some embodiments, the method has a sensitivity above 80%. In some embodiments, the method has an ROC greater than 70%. In some embodiments, the method has an AUC greater than 70%. In some embodiments, the method has a positive prediction value above 70%. In some embodiments, the method has a negative predicted value above 70%. In some embodiments, the reference gene performance profile is from subjects in a reference population of patients and / or healthy volunteers. In some embodiments, the comparing step includes at least one of: comparing digital images of performance profiles and comparing a database of performance data. In some embodiments of any of the above methods, the plasmablast biomarker is IgJ. In some embodiments of any of the above methods, the plasmablast biomarker is Mzb1. In some embodiments of any of the above methods, the plasmablast biomarker is Txndc5. In some embodiments of any of the above methods, the one or more plasmablast biomarkers are IgJ and Mzb1. In some embodiments of any of the above methods, the one or more plasmablast biomarkers are IgJ and Txndc5. In some embodiments of any of the above methods, the one or more plasmablast biomarkers are Txndc5 and Mzb1. In some embodiments of any of the above methods, the one or more plasmablast biomarkers are IgJ, Mzb1, and Txndc5. In some of the above embodiments, the sample is a urine sample. In some embodiments, the sample is a blood sample. In some embodiments, the biological sample is selected from the group consisting of blood, serum, plasma, and peripheral blood mononuclear cells (PBMC). In some embodiments, the biological sample is RNA obtained from blood, such as whole blood or a cell segment of blood, such as PBMC. In some embodiments, the biological sample is serum or plasma. Samples can be taken before, during or after treatment. Samples can be obtained from patients who are suspected of or diagnosed with SLE or other immune or inflammatory diseases and therefore may require treatment. Alternatively, the sample may be obtained from a normal individual who is not suspected of having any disease. In some embodiments, RNA is extracted from a biological sample described herein prior to detecting or measuring the mRNA content of the marker. The presence and / or expression content / quantity of a biomarker can be determined qualitatively and / or quantitatively based on any suitable criteria known in the art, including (but not limited to) DNA, mRNA, cDNA, protein, protein fragments, and / Or the number of gene copies. In some embodiments, the presence and / or expression content / amount of the biomarker in the first sample is increased compared to the presence / absence and / or expression content / amount in the second sample. In certain embodiments, the presence / absence and / or performance content / amount of the biomarker in the first sample is reduced compared to the presence and / or performance content / amount in the second sample. In some embodiments, the second sample is a reference sample, a reference cell, a reference tissue, a control sample, a control cell, or a control tissue. Described herein are additional disclosures for determining the presence / absence and / or expression content / amount of a gene. In some embodiments of any of the methods, high performance refers to comparison to a reference sample, reference cell, reference tissue, control sample, control cell, or control tissue by methods known in the standard art, such as those described herein They are described) 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80% of the biomarker (e.g., protein or nucleic acid (e.g., gene or mRNA)) detected The overall increase is any one of%, 90%, 95%, 96%, 97%, 98%, 99% or more. In certain embodiments, high performance refers to an increase in the expression content / amount of a biomarker in a sample, where the increase is a corresponding biomarker in a reference sample, a reference cell, a reference tissue, a control sample, a control cell, or a control tissue The expression content / amount is at least about 1.5 ×, 1.75 ×, 2 ×, 3 ×, 4 ×, 5 ×, 6 ×, 7 ×, 8 ×, 9 ×, 10 ×, 25 ×, 50 ×, 75 × or Any of 100 ×. In some embodiments, high performance refers to about 1.5 times, about 1.75 times, about 2 times compared to a reference sample, reference cell, reference tissue, control sample, control cell, control tissue, or internal control (e.g., a housekeeping gene) , About 2.25 times, about 2.5 times, about 2.75 times, about 3.0 times, or about 3.25 times the overall increase. In some embodiments of any of the methods, reduced performance refers to a comparison of a reference sample, reference cell, reference tissue, control sample, control cell, or control tissue by a method known in the standard, such as Those described) are about 10%, 20%, 30%, 40%, 50%, 60%, 70%, biomarker (e.g., protein or nucleic acid (e.g., gene or mRNA)) detected, The overall reduction is any of 80%, 90%, 95%, 96%, 97%, 98%, 99% or more. In some embodiments, the reduced performance refers to a decrease in the expression content / amount of the biomarker in the sample, wherein the decrease is the performance of the corresponding biomarker in the reference sample, reference cell, reference tissue, control sample, control cell, or control tissue. The content / amount is at least about 0.9 ×, 0.8 ×, 0.7 ×, 0.6 ×, 0.5 ×, 0.4 ×, 0.3 ×, 0.2 ×, 0.1 ×, 0.05 ×, or 0.01 ×. The presence and / or performance content / amount of various biomarkers in a sample can be analyzed by a number of methods, many of which are known in the art and familiar to those skilled in the art, including (but not limited to) immunohistochemistry ( (`` IHC ''), Western blot analysis, immunoprecipitation, molecular binding analysis, ELISA, ELIFA, fluorescence activated cell sorting (`` FACS ''), MassARRAY, proteomics research, blood-based quantitative analysis ( (E.g., serum ELISA), biochemical enzyme activity analysis, in situ hybridization, Southern analysis, Northern analysis, whole genome sequencing, polymerase chain reaction (`` PCR '') (including quantitative real-time PCR (`` qRT-PCR ") and other amplification-type detection methods, such as branched-chain DNA, SISBA, TMA, and the like), RNA-Seq, FISH, microarray analysis, gene performance profiling, and / or continuous analysis of gene performance ("SAGE") and any of a variety of analyses that can be performed by protein, gene, and / or tissue array analysis. Typical protocols for assessing the status of genes and gene products are found in, for example, Ausubel et al., 1995, Current Protocols In Molecular Biology, Units 2 (Northern Blotting), 4 (Southern Blotting), 15 (Immunoblotting), and 18 (PCR Analysis). Multiplex immunoassays may also be used, such as those available from Rules Based Medicine or Meso Scale Discovery ("MSD"). In some embodiments, the presence and / or expression content / amount of a biomarker is determined using a method comprising: (a) performing a gene performance profiling on a sample (such as a subject's cancer sample), PCR (such as rtPCR), RNA-seq, microarray analysis, SAGE, MassARRAY technology or FISH; and b) determine the presence and / or performance content / amount of biomarkers in the sample. In some embodiments, a microarray method includes using a microarray wafer having one or more nucleic acid molecules that can hybridize to a nucleic acid molecule encoding the gene described above under stringent conditions, or having a nucleic acid molecule that can bind to the gene encoded by the gene. One or more polypeptides (such as peptides or antibodies) of one or more proteins. In one embodiment, the PCR method is qRT-PCR. In one embodiment, the PCR method is multiplex PCR. In some embodiments, gene performance is measured by a microarray. In some embodiments, gene performance is measured by qRT-PCR. In some embodiments, gene performance is measured by multiplex PCR. Methods for assessing mRNA in cells are well known and include, for example, hybridization analysis using complementary DNA probes (such as in situ hybridization using labeled RNA probes specific for one or more genes, northern blots And related technologies) and various nucleic acid amplification analyses (such as RT-PCR using complementary primers specific for one or more of the genes, and other amplification type detection methods, such as branched-chain DNA, SISBA, TMA, and Its analogs). Samples from mammals may be suitably analyzed for mRNA using northern, spot blotting, or PCR analysis. In addition, these methods may include one or more steps that allow the determination of the content of a target mRNA in a biological sample (e.g., by simultaneously checking the content of a comparative control mRNA sequence of a "housekeeper" gene such as a member of the actin family) ). Optionally, the sequence of the amplified target cDNA can be determined. Optional methods include protocols for examining or detecting mRNAs, such as target mRNAs, in tissue or cell samples by microarray technology. Using nucleic acid microarrays, test and control mRNA samples from test and control tissue samples are reverse transcribed and labeled to generate cDNA probes. The probe is then hybridized to a nucleic acid array immobilized on a solid support. The array is configured so that the sequence and position of the members of the array are known. For example, a series of genes whose performance is related to the clinical benefit of increased or decreased anti-angiogenesis therapy can be arranged on a solid support. Hybridization of a labeled probe with a particular array member indicates that a sample of the derived probe expresses that gene. According to some embodiments, the presence and / or expression content / amount is measured by observing the protein expression content of the aforementioned genes. In certain embodiments, the method comprises contacting a biological sample with an antibody of a biomarker as described herein under conditions that allow binding of the biomarker, and detecting whether a complex is formed between the antibody and the biomarker. This method can be an in vitro or in vivo method. In one embodiment, the antibody is used to select a subject suitable for therapy with a BTK inhibitor, such as to select an individual's biomarker. In certain embodiments, IHC and staining protocols are used to detect the presence and / or performance content / amount of biomarker proteins in a sample. IHC staining of tissue sections has been shown to be a reliable method for determining or detecting the presence of proteins in a sample. In some embodiments of any of the methods, assays, and / or sets, the plasmablast biomarker is selected from one or more of IgJ, Mzb1, and Txndc5. In some embodiments, IgJ, Mzb1 and / or Txndc5 are detected by immunohistochemistry. In some embodiments, the high performance of plasmablast biomarkers in samples from individuals is high protein performance, and in other embodiments the IHC assay is used. In one embodiment, the performance content of a biomarker is determined using a method comprising: (a) performing an IHC analysis on a sample having an antibody; and b) determining the performance content of a biomarker in the sample. In some embodiments, IHC staining intensity is determined relative to a reference. In some embodiments, the reference is a reference value. In some embodiments, the reference is a reference sample (eg, a control cell line stained sample). In some embodiments, the tissue is kidney tissue. In other embodiments, the above technique is performed using fluorescence in situ hybridization instead of IHC. IHC can be performed in combination with other techniques, such as morphological staining and / or fluorescent in situ hybridization. Two IHC methods are available; direct analysis and indirect analysis. According to the first analysis, the binding of the antibody to the target antigen was directly determined. This direct analysis uses labeled reactants, such as fluorescently labeled or enzyme-labeled primary antibodies, which can be visually inspected without the need for additional antibody interactions. In a typical indirect analysis, the unbound primary antibody is bound to the antigen and then the labeled secondary antibody is bound to the primary antibody. When the secondary antibody is bound to the enzyme label, a chromogenic or fluorescent substrate is added to provide visual inspection of the antigen. Because several secondary antibodies can react with different epitopes on the primary antibody, signal amplification occurs. Primary and / or secondary antibodies for IHC will usually be labeled by a detectable moiety. Several markers are available, which can be broadly grouped into the following categories: (a) Radioisotopes, such as35 S,14 C,125 I,3 H and131 I; (b) colloidal gold particles; (c) fluorescent labels, including (but not limited to) rare earth chelates (fluorene chelates), Texas Red, rhodamine , Fluorescein, dansyl, Lissamine, umbelliferone, phycocrytherin, phycocyanin or commercially available fluorescent groups, Derivatives such as SPECTRUM ORANGE7 and SPECTRUM GREEN7 and / or any one or more of the above; (d) various enzyme substrates are labeled as available signatures and US Patent No. 4,275,149 provides a review of some of these. Examples of enzyme labels include luciferase (e.g., firefly luciferase and bacterial luciferase; U.S. Patent No. 4,737,456), luciferin, 2,3-dihydrophthalazinedione, malate Catalase, urease, peroxidase (such as horseradish peroxidase (HRPO)), alkaline phosphatase, β-galactose, amylase, lysozyme, sugar oxidase (e.g., glucose oxidase, Galactose oxidase and glucose-6-phosphate dehydrogenase), heterocyclic oxidases (such as urase and xanthine oxidase), lactoperoxidase, microperoxidase and the like. Examples of the enzyme substrate combination include, for example, horseradish peroxidase (HRPO) having a hydroperoxidase as a substrate; alkaline phosphatase (AP) having p-nitrophenyl phosphate as a color substrate; and Β-D-galactose (for example, p-nitrophenyl-β-D-galactose) or fluorescein (for example, 4-methylumbellyl-β-D-galactose) β-D-Gal). For a general review of these, see US Patent Nos. 4,275,149 and 4,318,980. In some embodiments of any of the methods, a plasmablast biomarker is detected by immunohistochemistry using a diagnostic antibody (ie, a primary antibody). In some embodiments, the tissue to be analyzed is kidney tissue. In some embodiments, the diagnostic antibody specifically binds IgJ, Mzb1, or Txndc5. In some embodiments of any of the diagnostic antibodies, the diagnostic antibody is a non-human antibody. In some embodiments, the diagnostic antibody is a rat, mouse, or rabbit antibody. In some embodiments, the diagnostic antibody is a monoclonal antibody. In some embodiments, the diagnostic antibody is directly labeled. In an alternative method, a sample can be contacted with an antibody specific for that biomarker under conditions sufficient to form an antibody biomarker complex, and the complex can then be detected. The presence of biomarkers can be detected in a variety of ways, such as by Western blotting and ELISA procedures for analyzing a wide variety of tissues and samples, including plasma or serum. A wide range of immunoassay techniques using this type of analysis are available, see, for example, U.S. Patent Nos. 4,016,043, 4,424,279, and 4,018,653. These analyses include non-competitive single-point and two-point or "sandwich" analysis, as well as traditional competitive combined analysis. These analyses also include direct binding of labeled antibodies to target biomarkers. Functional or activity-type analysis can also be used to detect the presence and / or expression content / amount of selected biomarkers in a tissue or cell sample. For example, if the biomarker is an enzyme, we can perform an analysis known in the art to determine or detect the presence of a given enzyme activity in a tissue or cell sample. In certain embodiments, the sample is normalized for both the difference in the amount of biomarker analyzed and the variability in the mass of the sample used and the variability between the analyses run. This standardization can be achieved by detecting and incorporating the performance of certain standardized biomarkers, including well-known housekeeping genes such as ACTB. Alternatively, normalization may be based on the average or intermediate signals of all analyzed genes or a larger subset of them (a fully normalized approach). On a gene-by-gene basis, the measured normalized amount of mRNA or protein in an individual sample is compared with the amount found in the reference set. The normalized expression content for each mRNA or protein / test sample / subject can be expressed as a percentage of the expression content measured in the reference set. The presence and / or performance content / amount measured in a particular subject sample to be analyzed will be reduced by a few percentage points within this range, which can be determined by methods well known in the art. In some embodiments, the relative expression content of a gene is determined as follows: relative expression gene 1 sample 1 = 2 exp (Ct housekeeper gene-Ct gene 1) and Ct measured in the sample. Relative expression gene 1 reference RNA = 2 exp (Ct housekeeping gene-Ct gene 1) and Ct measured in the reference sample. Normalized relative expression gene 1 sample 1 = (relative expression gene 1 sample 1 / relative expression gene 1 reference RNA) × 100 Ct is a threshold cycle. Ct is the number of cycles, where the fluorescence generated in the reaction exceeds the threshold line. All experiments were standardized as reference RNA, which is a comprehensive mix of RNA from various tissue sources (eg, reference RNA # 636538 from Clontech, Mountain View, CA). The same reference RNA is included in each qRT-PCR run, allowing comparison of results between different experimental runs. In one embodiment, the sample is a clinical sample. In another embodiment, the sample is used for diagnostic analysis. In some embodiments, the sample is obtained from a tissue. Tissue biopsies are often used to obtain representative tissue fragments. Alternatively, tumor cells can be obtained indirectly in the form of tissues or fluids which are known or considered to contain the cells of interest. Genes or gene products can be detected from tissues or from other body samples such as urine, sputum, serum or plasma. By screening these body samples, it is easier to monitor the progress of the therapy by testing these body samples for the target gene or gene product. In certain embodiments, the reference sample, reference cell, reference tissue, control sample, control cell, or control tissue is a single sample or multiple samples from the same subject or individual combination, in addition to obtaining one of the test samples The multiple samples are obtained at multiple different time points. For example, a reference sample, a reference cell, a reference tissue, a control sample, a control cell, or a control tissue is obtained from the same subject or individual at an earlier point in time than the test sample is obtained. This reference sample, reference cell, reference tissue, control sample, control cell, or control tissue may be useful if a reference sample is obtained during the initial diagnosis of the disease and a test sample is subsequently obtained as the disease progresses. In certain embodiments, the reference sample, reference cell, reference tissue, control sample, control cell, or control tissue is a merged plurality of samples from one or more healthy individuals who are not the subject or individual. In certain embodiments, the reference sample, reference cell, reference tissue, control sample, control cell, or control tissue is a combined number of individuals from one or more individuals with a disease or disorder that is not the subject or individual. Samples. In certain embodiments, the reference sample, reference cell, reference tissue, control sample, control cell, or control tissue is a pooled RNA sample from normal tissue that is not a subject or individual or one or more individuals After pooling plasma or serum samples. In certain embodiments, the reference sample, reference cell, reference tissue, control sample, control cell, or control tissue is from a pooled RNA sample from a tissue that is not a subject or individual or from one with a disease or disorder Pooled plasma or serum samples from one or more individuals. In certain embodiments, the reference sample, reference cell, reference tissue, control sample, control cell, or control tissue is a sample cell line. In certain embodiments, the reference sample, reference cell, reference tissue, control sample, control cell, or control tissue is blood. In some embodiments, the sample is a tissue sample from an individual. In some embodiments, the tissue sample is a blood or urine sample. In some embodiments, the tissue sample is a blood sample. In some embodiments of any of the methods, the BTK inhibitor is a small molecule BTK inhibitor. In some embodiments, the small molecule BTK inhibitor is Compound (A) or a pharmaceutically acceptable salt thereof. In some embodiments of any of the methods, the individual or patient according to any of the above embodiments may be a human. In another embodiment, provided herein are methods of treating SLE. In one embodiment, the method comprises administering to a subject having SLE an effective amount of a small molecule BTK inhibitor. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, as described below. In some embodiments, the individual may be a human. The BTK inhibitors described herein can be used alone or in combination with other agents in therapy. For example, the additional therapeutic agent may be an anti-inflammatory agent, an immunomodulator, a chemotherapeutic agent, an apoptosis enhancer, a neurotrophic factor, an agent for treating cardiovascular diseases, an agent for treating liver diseases, an antiviral agent, or an agent for treating blood disorders. 5. Medicines for treating diabetes and medicines for immunodeficiency disorders. The second therapeutic agent may be an NSAID anti-inflammatory agent. The second therapeutic agent may be a chemotherapeutic agent. The second compound of the pharmaceutical combination formulation or dosing regimen preferably has the complementary activity of compound (I) so that they do not adversely affect each other. In some embodiments, the additional therapeutic agent is selected from the group consisting of a corticosteroid (eg, prednisone, prednisolone, methylprednisolone, and hydrocortisone); Disease-modifying antirheumatic drugs ("DMARD", such as immunosuppressive or anti-inflammatory agents); antimalarials (such as hydroxychloroquine and chloroquine); immunosuppressants (such as cyclophosphamide, azathioprine, mycophenol Mycophenolate mofetil, methotrexate); anti-inflammatory agents (e.g., aspirin, NSAID (e.g., ibuprofen, naproxen, indole) Indomethacin, nabumetone, celecoxib); antihypertensive agents (e.g., calcium channel blockers (e.g., amlodipine, nifedipine (e.g. nifedipine)) and diuretics (for example, furosemide)); statins (for example, atorvastatin, fluvastatin, lovastatin, lovastatin, pill Pitavastatin, pravastatin, rosuvastat in) and simvastatin); anti-B cell agents (e.g., anti-CD20 (e.g., rituximab), anti-CD22); anti-B lymphocytic stimulants ("anti-BLyS", e.g., belizumab) (belimumab, blisibimod)); type 1 interferon receptor antagonists (e.g., anifrolumab); T cell modulators (e.g., rigerimod); Abatacept; anticoagulants (eg, heparin, warfarin); and vitamin D supplements. Combination therapies can be administered simultaneously or in a sequential regimen. When administered sequentially, the combination can be administered in two or more administration forms. Combination administration includes co-administration using separate formulations or a single pharmaceutical formulation, and continuous administration in any order, with two (or all) active agents preferably exhibiting their biological activity simultaneously over a period of time. A suitable dose of any of the above co-administered medicaments is the currently used dose and can be reduced due to the combined effect (synergy) of the additional therapeutic agent. Combination therapies can be compounded such that the effects obtained when the active ingredients are used together are greater than the sum of the effects produced by the compounds used alone. Synergistic effects can be obtained when the active ingredients are: (1) simultaneously administered or delivered; (2) alternately or simultaneously administered; or (3) by some other protocol. When delivered in alternating therapies, synergistic effects can be obtained when the compound is continuously administered or delivered. In general, an effective dose of each active ingredient is administered sequentially (i.e., continuously) during alternation therapy, whereas in combination therapy, an effective dose of two or more active ingredients is administered together. In combination therapy, the kit may include (a) a first container having the dosage form composition of the present invention and, as appropriate, (b) a second pharmaceutical formulation contained therein for use with the dosage form composition of the present invention. Co-administered second container. In such aspects, the kit may include a container for containing a separate composition, such as a split bottle or a split foil pack, however, the separate composition may also be contained in a single non-separated container. Generally, the kit contains instructions for administering the individual components. The kit form is particularly useful when the individual components are preferably administered in different dosage forms (e.g., orally and parenterally), when administered at different dosing intervals, or when the prescriber needs to titrate the individual components of the combination. advantageous. BTK inhibitors can be administered by any suitable means, including oral, parenteral, intrapulmonary, and intranasal, and intralesional administration for local treatment as needed. In a preferred embodiment, a BTK inhibitor is administered orally. Oral dosage forms comprising a BTK inhibitor include, but are not limited to, lozenges or capsules comprising a BTK inhibitor or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients. In some embodiments, lozenges or capsules comprising a BTK inhibitor can be administered once or twice daily according to the methods provided herein. In certain embodiments provided herein, the oral dosage form is a lozenge comprising Compound (A) or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal or subcutaneous administration. Depending on the short-term or long-term nature of the administration, it may be administered by any suitable route (e.g., by injection, such as intravenous or subcutaneous). Various dosing schedules are covered herein, including (but not limited to) a single administration or multiple administrations at different time points, rapid administration, and pulsed infusions. The BTK inhibitors described herein can be formulated, administered, and administered in a manner consistent with good medical practice. In this context, considerations include the specific disease or condition being treated, the specific mammal being treated, the clinical condition of the individual patient, the cause of the disease or condition, the site of delivery of the agent, the method of administration, and the timing of administration And other factors known to medical practitioners. BTK inhibitors need not be, but are optionally formulated with one or more agents currently used to prevent or treat a disease or disorder. The effective amount of these other agents depends on the amount of BTK inhibitor present in the formulation, the type of disease or disorder or treatment, and other factors as discussed above. These agents are typically used at the same dose and in the route of administration as described herein, or at about 1% to 99% of the dose described herein, or at any dose and empirically / clinically determined as appropriate Any way to use. III. contain BTK Inhibitor Therapeutic composition The compositions, methods, and kits herein provide small molecule BTK inhibitors. The small molecule BTK inhibitors provided herein are preferably organic molecules other than binding polypeptides or antibodies, and can be identified and chemically synthesized using known methods. Binding organic small molecules are typically smaller than about 2000 Daltons, and alternatively smaller than about 1500, 750, 500, 250, or 200 Daltons, where it is possible to bind (preferably, specifically) a BTK as described herein. These small organic molecules can be identified using well-known techniques without undue experimentation. In this regard, it should be noted that techniques for screening small organic libraries of molecules that can be bound to polypeptide targets are well known in the art (see, for example, PCT Publication Nos. WO 2000/00823 and WO 2000 / 39585). Binding organic small molecules can be, for example, aldehydes, ketones, oximes, amidines, semi-carbamidines, hydrazines, primary amines, secondary amines, tertiary amines, N-substituted hydrazines, hydrazine, alcohols, ethers, thiols, thioether , Disulfide, carboxylic acid, ester, ammonium, urea, carbamate, carbonate, ketal, thioketal, acetal, thioacetal, aryl halide, aryl sulfonate, alkyl Halides, alkyl sulfonates, aromatic compounds, heterocyclic compounds, anilines, olefins, alkynes, glycols, amino alcohols, oxazolines, oxazolines, thiazolines, thiazolines, enamines, sulfonium Amines, epoxides, aziridines, isocyanates, sulfonyl chlorides, diazo compounds, acid chlorides or the like. In some embodiments of any of the methods, the BTK inhibitor is selected from the group consisting of: ibrutinib, acalabrutinib, spebrutinib, BIIB068 (Biogen), BMS-986195 (Bristol-Myers Squibb), BMS-986142 (Bristol-Myers Squibb), BMS-935177 (Bristol-Myers Squibb), M2951 (Merck KGaA), PRN-1008 (Principia Biopharma), HM71224 / LY3337641 (Hanmi / Lilly), ONO-4059 / GS-4059 (Gilead / Ono), AC0058 (ACEA Biosciences), AC0025 (ACEA Biosciences), ABBV-599 (AbbVie), ABBV-105 (AbbVie), PF-303 (Pfizer), BI-BTK1 (Boehringer Ingelheim), CC90008 (Celgene), AS550 (Carna Biosciences), ARQ 531 (Arqule), AEG42766 (Aegera Therapeutics), BGB-3111 (Beigene), RN486 (Simcere Pharma), HCI- 1401 (LSK BioPharma / Hustman Cancer Inst.), KBP-7536 (KBP Bioscience), RDX002 (RedX Biopharma), SNS-062 (Sunesis), TAS5315 (Taiho Pharma), TAX-020 (Takeda), WX486 / WXFL-10230486 (WuXi AppTec / Humanwell) and X-022 (X-Rx Discovery). In some embodiments, the BTK inhibitor is Compound (A) or a pharmaceutically acceptable salt thereof. The pharmaceutically acceptable salts of the BTK inhibitors provided herein can be used in the methods herein. As used herein, the term "pharmaceutically acceptable salt" is intended to include salts of active compounds prepared with relatively non-toxic acids or bases, depending on the particular substituents present on the compounds described herein. When the compound of the present invention contains a relatively acidic functional group, a base addition salt can be obtained by contacting the neutral form of such a compound with a sufficient amount of the desired base in the absence of a solvent or in a suitable inert solvent. Examples of salts derived from a pharmaceutically acceptable inorganic base include aluminum salts, ammonium salts, calcium salts, copper salts, iron salts, ferrous salts, lithium salts, magnesium salts, manganese salts, manganese salts, potassium salts, Sodium, zinc and similar salts. Salts derived from pharmaceutically acceptable organic bases include salts of primary, secondary, and tertiary amines, including substituted amines, cyclic amines, naturally occurring amines, and the like, such as arginine, betaine , Caffeine, choline, N, N'-benzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethyl Morpholine, N-ethylpiperidine, reduced glucosamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methyl reduced glucosamine, morpholine, piperazine, Piperidine, polyamine resin, procaine, purine, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like. When the compounds of the present invention contain relatively basic functional groups, acid addition salts can be obtained by contacting the neutral form of these compounds with a sufficient amount of the desired acid in the absence of a solvent or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include their acid addition salts derived from inorganic acids, such as hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, monohydrocarbonic acid, phosphoric acid, monohydrophosphoric acid, Dihydrophosphoric acid, sulfuric acid, monohydrosulfuric acid, hydroiodic acid or phosphorous acid and similar acids; and salts derived from relatively non-toxic organic acids such as acetic acid, propionic acid, isobutyric acid, malonic acid, benzene Formic acid, succinic acid, suberic acid, fumaric acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-tolylsulfonic acid, citric acid, tartaric acid, methanesulfonic acid and the like. Also included are salts of amino acids such as arginine and similar acids, and salts of organic acids such as glucuronic acid or galacturonic acid and similar acids (see, for example, Berge, SM et al., "Pharmaceutical Salts", Journal of Pharmaceutical Science, 1977, 66, 1-19). Certain specific compounds of the present invention contain both basic and acidic functional groups that allow the compounds to be converted into base addition salts or acid addition salts. The compound neutral form can be regenerated by contacting the salt with a base or acid and isolating the parent compound in a conventional manner. The parent form of a compound differs from various salt forms in certain physical properties, such as solubility in polar solvents, but for the purposes of the present invention, in other respects, salts are equivalent to the parent form of the compound. In addition to salt forms, the present invention provides compounds in prodrug form. As used herein, the term "prodrug" refers to those compounds that readily undergo chemical changes under physiological conditions to provide the compounds of the invention. In addition, prodrugs can be converted to compounds of the invention in an ex vivo environment by chemical or biochemical methods. For example, when a current drug is placed in a transdermal patch reservoir with a suitable enzyme or chemical agent, it can be slowly converted into a compound of the invention. The prodrugs of the present invention include compounds in which the polypeptide chain of an amino acid residue or two or more (e.g., two, three, or four) amino acid residues is covalently attached to the present invention via an amidine or ester bond Free amine, hydroxyl, or carboxylic acid groups of the compound. Amino acid residues include (but are not limited to) 20 naturally occurring amino acids commonly represented by three-letter symbols, and also include phosphoselanine, threonine phosphate, tyrosine phosphate, and 4-hydroxyproline Amino acid, hydroxy lysine, chain lysine, iso-chain lysine, γ-carboxyglutamic acid, hippuric acid, octaindole-2-carboxylic acid, statine, 1, 2, 3, 4-tetrahydroisoquinoline-3-carboxylic acid, penicillamine, ornithine, 3-methylhistidine, n-valine, β-alanine, γ-aminobutyric acid, citrulline, high Cysteine, homoserine, methyl-alanine, p-benzylidenephenylalanine, phenylglycine, propargylglycine, sarcosine, methionine, and tertiary Butylglycine. Other types of prodrugs are also covered. For example, the free carboxyl group of a compound of the invention can be derived as amidamine or an alkyl ester. As another example, a compound of the present invention containing a free hydroxyl group can be converted by, for example, but not limited to, a phosphate, a hemisuccinate, a dimethylaminoacetate, or a phosphonium oxymethyloxy group. The carbonyl group is derived as a prodrug, as outlined in Fleisher, D. et al. (1996) Improved oral drug delivery: solubility limitations overcome by the use of prodrugs Advanced Drug Delivery Reviews, 19: 115. It also includes hydroxyl and amine carbamate prodrugs, like carbonate prodrugs, and also includes hydroxyl sulfonates and sulfates. It also covers hydroxy groups derived from (fluorenyl) methyl and (fluorenyl) ethyl ether, where fluorenyl may be optionally substituted with groups including, but not limited to, ether, amine, and carboxylic acid functional groups Alkyl esters, or amino esters in which the fluorenyl group is as described above. This type of prodrug is described in J. Med. Chem., (1996), 39:10. More specific examples include the replacement of a hydrogen atom of an alcohol group with a group such as (C1 -6 ) Alkoxymethyl, 1-((C1 -6 ) Alkanoyloxy) ethyl, 1-methyl-1-((C1 -6 ) Alkoxy) ethyl, (C1 -6 ) Alkoxycarbonyloxymethyl, N- (C1 -6 ) Alkoxycarbonylaminomethyl, succinyl, (C1 -6 ) Alkyl group, α-amino group (C1 -4 ) Alkyl, aryl, and α-aminofluorenyl or α-aminofluorenyl-α-aminofluorenyl, wherein each α-aminofluorenyl is independently selected from naturally occurring L-amino acids , P (O) (OH)2 , -P (O) (O (C1 -6 )alkyl)2 Or glycosyl (a group resulting from removal of a hydroxyl group in the hemiacetal form of a carbohydrate). For additional examples of prodrug derivatives, see, for example, a) Design of Prodrugs, edited by H. Bundgaard, (Elsevier, 1985) and Methods in Enzymology, vol. 42, pp. 309-396, edited by K. Widder et al. (Academic Press, 1985); b) A Textbook of Drug Design and Development, edited by Krogsgaard-Larsen and H. Bundgaard, Chapter 5 "Design and Application of Prodrugs", edited by H. Bundgaard, pp. 113-191 ( 1991); c) H. Bundgaard, Advanced Drug Delivery Reviews, 8: 1-38 (1992); d) H. Bundgaard, et al., Journal of Pharmaceutical Sciences, 77: 285 (1988); and e) N. Kakeya et al., Chem. Pharm. Bull., 32: 692 (1984), each of which is specifically incorporated herein by reference. Certain compounds of the invention may exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, solvated forms are equivalent to unsolvated forms and are intended to be encompassed within the scope of the present invention. Certain compounds of the invention may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent to the uses covered by the present invention and are intended to be within the scope of the present invention. Certain compounds of the invention have asymmetric carbon atoms (optical centers) or double bonds; racemates, diastereomers, geometric isomers, regioisomers, and individual isomers (e.g., individual Enantiomers) are intended to be included within the scope of this invention.IV. Pharmaceutical formulations Pharmaceutical formulations of BTK inhibitors are provided in the methods and kits herein. In some embodiments of any of the methods, from about 0.1 mg / kg / day to about 100 mg / kg / day, about 0.5 mg / kg / day to about 20 mg / kg / day, based on the patient's weight, Or, a BTK inhibitor (for example, compound (A) or a pharmaceutically acceptable salt thereof) is administered at a dose of about 1 mg / kg / day to about 10 mg / kg / day. In some embodiments, Compound (A) or a pharmaceutically acceptable salt thereof is administered as a lozenge in a dose of about 10 to 800 mg. In some embodiments, Compound (A) is administered as a free base in a lozenge at a dose of about 25 to 300 mg. In some embodiments, the lozenge contains 25 to 300 mg of compound (A) as a free base and fumaric acid, wherein the weight ratio of compound (A) to fumaric acid is about 1: 5 to about 3: 1; or about 1: 2 to about 2: 1; or about 1: 1.5 to about 1.5: 1. In some embodiments, the lozenge comprises 25 to 300 mg of compound (A) as a free base and fumaric acid, and wherein the fumaric acid content is from about 5 wt% to about 50 wt%, about 5 Wt% to about 40 wt%, about 5 wt% to about 30 wt%, about 10 wt% to about 30 wt%, about 20 wt% to about 25 wt%, about 5 wt% to about 15 wt%, or about 10 % By weight to about 15% by weight. In some of the above embodiments, the tablet weight is about 100 mg, about 200 mg, about 300 mg, about 400 mg, about 500 mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg, or about 1000 mg. In some embodiments, the lozenge further comprises at least one pharmaceutically acceptable excipient selected from the group consisting of a filler, a binding agent, a disintegrant, a lubricant, and a sliding agent. In some embodiments, the lozenge comprises lactose and microcrystalline cellulose. The tablet composition of the present invention may further suitably include one or more pharmaceutically acceptable excipients selected from, but not limited to, fillers (diluents), disintegrants, binders, slip agents, and lubricants. Agent. Fillers (or diluents) can be used to increase the total volume of the powdered medicine that makes up the lozenge. Disintegrants can be used to promote the disintegration of lozenges into smaller pieces when ingested, ideally individual drug particles, and thereby facilitate rapid dissolution and absorption of the drug. Binders can be used to ensure that the granules and lozenges can be formed to have the required mechanical strength and to hold the lozenges together after they have been compressed, preventing them from disintegrating into their component powders during packaging, delivery and normal operations. Slip agents can be used to improve the flowability of the powders that make up the tablets during production. Lubricants can be used to ensure that the ingot powder does not stick to the equipment used to press the tablets during manufacturing, to improve the flow of the powder during mixing and pressing, and to minimize friction and cracking when the final tablets are sprayed from the device. Fillers and binders may include dibasic calcium phosphate, microcrystalline cellulose (Avicel®), lactose or any other suitable bulking agent. Examples of suitable fillers include microcrystalline cellulose, such as Avicel PH 101, Avicel PH102, Avicel PH 200, Avicel PH 105, Avicel DG, Ceolus KG 802, Ceolus KG 1000, SMCCSO, and Vivapur 200; lactose monohydrate, such as lactose FastFlo ; Co-processed microcrystalline cellulose with other excipients, such as co-processed microcrystalline cellulose with lactose monohydrate (MicroceLac 100) and co-processed with colloidal silica (SMCCSO, Prosolv 50, and Prosolv HD 90) Microcrystalline cellulose; mixtures of isomaltulose derivatives, such as galenIQ; and other suitable fillers and combinations thereof. The filler may be in the form of an intragranular component and / or an extragranular component. In some specific aspects, the lozenge composition of the present invention comprises lactose and microcrystalline cellulose. A disintegrant may be included in the disclosed formulation to promote the separation of the particles within the pressed product from each other and to maintain the released particles from each other. The disintegrant may be present as an intragranular component and / or an extragranular component. The disintegrant may include any suitable disintegrant such as a cross-linked polymer such as cross-linked polyvinyl pyrrolidone and cross-linked sodium carboxymethyl cellulose or cross-linked sodium carboxymethyl cellulose. In some specific aspects, the disintegrant is croscarmellose sodium. The disintegrant content is suitably about 1% by weight, about 1.5% by weight, about 2% by weight, about 2.5% by weight, about 3% by weight, about 3.5% by weight, about 4% by weight, about 4.5% by weight, or about 5% by weight. Weight percent and ranges thereof, such as about 1 weight percent to about 5 weight percent or about 2 weight percent to about 4 weight percent. Slip agents may include, for example, colloidal silica, including highly dispersed silica (Aerosil®) or any other suitable slip agent, such as animal or vegetable fats or waxes. In some specific aspects, the sliding agent is fumed silica. The slip agent content is suitably about 0.1% by weight, about 0.5% by weight, about 1% by weight, about 1.5% by weight, about 2% by weight, about 2.5% by weight, or about 3% by weight, and ranges thereof, such as about 0.1% by weight To about 3% by weight, about 0.5% to about 2% by weight, and about 0.5% to about 1.5% by weight. Lubricants can be used to compact particles in pharmaceutical compositions. Lubricants may include, for example, polyethylene glycol (e.g., a molecular weight of about 1000 to about 6000), magnesium and calcium stearate, sodium stearyl fumarate, talc, or any other suitable lubricant. In some specific aspects, the lubricant is magnesium stearate and / or sodium stearyl fumarate. The lubricant may be in the form of an intragranular component and / or an extragranular component. The lubricant content is suitably about 0.5% by weight, about 1% by weight, about 1.5% by weight, about 2% by weight, about 2.5% by weight, about 3% by weight, about 3.5% by weight, about 4% by weight, and about 4.5% by weight Or about 5 wt% and ranges thereof, such as about 0.5 wt% to about 5 wt%, about 1 wt% to about 4 wt%, about 1 wt% to about 3 wt%, or about 1 wt% to about 2 wt%. A coating, such as a film coating, may be applied to the lozenges of the invention. Film coatings can be used, for example, to help lozenges be easily swallowed. Film coating can also be used to improve taste and appearance. If desired, the film coating may be an enteric coating. The film coating may include polymerizable film-forming materials such as hydroxypropyl methyl cellulose, hydroxypropyl cellulose, acrylate or methacrylate copolymers, and polyvinyl alcohol-polyethylene glycol graft copolymers such as Opadry and Kollicoat IR. In addition to the film-forming polymer, the film coating may additionally contain plasticizers, such as polyethylene glycol; surfactants, such as Tween® type; and optionally pigments, such as titanium dioxide or iron oxide. The film coating may also contain talc as an anti-adhesive agent. Film coatings typically comprise less than about 5% by weight of the dosage form. The formulations herein may also contain more than one active ingredient necessary for the particular indication being treated, preferably those active ingredients having complementary activities that do not adversely affect each other. Such active ingredients are suitably present in combination in amounts effective to achieve the intended purpose. The active ingredients can be coated in the prepared microcapsules, for example, by coacervation technology or by interfacial polymerization, such as hydroxymethyl cellulose or gelatin microcapsules and poly (methyl methacrylate) microcapsules, respectively, in a gel. Drug delivery systems (eg, liposomes, albumin microspheres, microemulsions, nanoparticle and nanocapsules) or macroemulsions. These techniques are revealed inRemington's Pharmaceutical Sciences 16th ed., Osol, A. Ed. (1980). Delayed-release preparations can be prepared. Suitable examples of extended release formulations include semi-permeable matrices of solid hydrophobic polymers containing BTK inhibitors, which matrices are in the form of shaped articles, such as films or microcapsules. Formulations for in vivo administration are generally sterile. Sterility can be easily achieved by, for example, filtration through a sterile filtration membrane.V. product In another embodiment, an article of manufacture containing materials suitable for treating, preventing, and / or diagnosing the conditions described above is provided. Articles of manufacture include containers and markings or drug instructions on or associated with the containers. Suitable containers include, for example, bottles, vials, syringes, IV solution bags, and the like. The container can be formed from a variety of materials, such as glass or plastic. A container holds a composition itself or a combination of a composition and another composition that is effective in treating, preventing and / or diagnosing a condition, and may have a sterile access port (e.g., the container may have a needle Stopper bag or vial of intravenous solution). At least one active agent in the composition is a BTK inhibitor described herein. The indicia or the pharmaceutical label indicates that the composition is used to treat the condition of choice. In addition, the article of manufacture may comprise (a) a first container and a composition contained therein, wherein the composition comprises a BTK inhibitor; and (b) a second container and a composition contained therein, wherein the composition comprises another Cytotoxic or additional therapeutic agents. In some embodiments, the article of manufacture comprises a container, a label on the container, and a composition contained within the container; wherein the composition includes one or more reactants (bound to one or more biomarkers or described herein Primary antibodies to probes and / or primers of one or more of the biomarkers (eg, B-9 Santa Cruz Biotechnology antibodies), and a label on the container indicates that the composition can be used to evaluate one or more biomarkers in a sample Presence, and instructions for using a reagent for assessing the presence of one or more biomarkers in a sample. The article of manufacture may further include a set of instructions and materials for preparing a sample and utilizing a reagent. In some embodiments, the article of manufacture may include a reactant, such as both a primary antibody and a secondary antibody, wherein the secondary antibody is bound to a label, such as an enzyme label. In some embodiments, an article, one or more probes and / or primers directed to one or more of the biomarkers described herein. The article of manufacture in this embodiment may further include a pharmaceutical instruction sheet indicating that the composition can be used to treat a particular condition. Alternatively or in addition, the article of manufacture may further comprise a second (or third) container containing a pharmaceutically acceptable buffer such as bacteriostatic water for injection (BWFI), phosphate buffered saline, Ringer's solution (Ringer's solution) and dextrose solution. It may further include other materials required from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes. Other optional components in the preparation include one or more buffers (e.g., blocking buffer, wash buffer, substrate buffer, etc.), other reagents (such as substrates that are chemically altered by enzyme labeling) ) (Such as chromogens), epitope recovery solutions, control samples (positive and / or negative controls), control sections, and the like.Examples The following are examples of methods and compositions. It should be understood that various other embodiments may be implemented in view of the general description provided above.Blood sample analysis : 3- Analysis of Gene Plasma Cell Tag Performance. Blood was collected in a PAXgene RNA test tube (PreAnalytiX); total RNA was extracted using a commercially available kit according to the manufacturer's instructions (Qiagen).Biomarkers: IgJ, TXNDC5, MZB1.Reference gene: TMEM55B uses the Human Genome U133 Plus 2.0 array (Affymetrix Inc., Santa Clara, CA) to evaluate the performance of candidate biomarker genes in blood samples. Microarray hybridization was performed by Asuragen Inc. (Austin, TX). The original CEL archives were summed and standardized using Robust Multi-array Averaging (RMA) and analyzed using R and Bioconductor. Alternatively, the performance of candidate biomarker genes in blood samples is quantified by Fluidigm qPCR analysis. The three gene scores were calculated based on the average of IgJ, TXNDC5, and MZB1, and standardized using the reference gene TMEM55B. This analysis generated on the Cobas 4800 platform (Roche Molecular Systems) was used to evaluate blood samples.Examples 1 : Characteristics of plasmablast transcriptome Perform transcriptional analysis of CD20loCD38 + plasmablasts, CD20 + CD27 + activated B cells, and CD20 + CD27-native B cells differentiated in vitro to identify genes with strong differentiation among B cell subsets (Figure 1A-1) . At a false discovery rate (FDR) of 0.001, 86 genes were identified, and their performance in plasmablasts was> 10-fold higher than either of activated B cells or native B cells. Further refinement of these data was performed to include only genes with> 5 nRPKM in plasmablasts, resulting in a total of 40 genes. Many of these genes include heavy and light chain segments, as well as genes involved in the biosynthesis of immunoglobulin proteins. Biomarker candidates that were not part of the immunoglobulin locus were selected, so these candidates were removed from the candidate gene list (Figure 1B). It has been demonstrated that plasmablast differentiation is regulated by Bruton's tyrosine kinase (BTK) activity by performing an in vitro plasmablast differentiation assay, in which human memory B cells are placed under differentiation conditions and subsequently flow cytometry is used Cytometry quantifies CD20loCD38 ++ plasmablasts after 5 days. A specific and potent inhibitor of BTK kinase activity (GDC-0852) was used in a dose-dependent manner to inhibit CD40L-induced plasmablast differentiation (Figure 7). GDC-0852 is (S) -2- (5-fluoro-2- (hydroxymethyl) -3- (1-methyl-5- (5- (2-methyl-4- (oxetan) -3-yl) piperazin-1-yl) pyridin-2-ylamino) -6- pendantoxy-1,6-dihydropyridin-3-yl) -phenyl) -3,4,6, 7,8,9 hexahydropyrido [3,4-b] indolinazin-1 (2H) -one, the structure of which is shown below:. In order to ensure that the selected biomarker candidates can accurately determine the fraction of plasmablasts in samples with high sensitivity and specificity, 40,000 to 39 of plasmablasts derived in vitro from 3 donors are distinguished Each cell was subjected to a serial 2-fold dilution to 1,000,000 PBMCs derived from two separate donors. Fluidigm was used to assess the expression content of candidate plasmablast marker genes and was reported as ΔCt relative to the housekeeping gene HPRT1. Linear regression was used to model the ΔCt of candidate genes predicted by log10 plasmablast frequency, with plasmablast donors and PBMC donors as covariates. Most of our candidate genes show a strong correlation with plasmablast frequency and minimal differences between plasmablast donors. Three genes, IGJ, MZB1, and TXNDC5 performed particularly well, showing r of 0.84, 0.75, and 0.69, respectively2 (Figures 2B-1, 2B-2, 2B-3, 2B-4). Take the average of the three genes as the label score to generate r2 A label score of 0.79. To verify the genetic signature of differentiated plasmablasts in vivo, their performance was measured in plasmablasts sorted directly from healthy donors inoculated with influenza vaccinia from five weeks. All three candidate biomarker genes are more highly expressed in plasmablasts than both native and memory B cells (Figures 2C-1, 2C-2, 2C-3, 2C-4). Example 2: Plasmablast marker gene correlates with in vivo plasmablast frequency. Flow cytometry was used to measure the frequency of plasmablasts in whole blood in a lupus patient population from a ROSE phase II clinical trial [3]. , Which has paired RNA sequencing data. Selected Plasmablast Tag Genes Displayed IgD-CD19+ CD27++ CD38++ High correlation of plasmablast frequency (Figures 2D-1, 2D-2, 2D-3, 2D-4). IGJ, MZB1, and TXNDC5 showed the highest correlation coefficients with plasmablast content. Spearman correlation coefficients were 0.66, 0.71, and 0.71, respectively. The average of the three tag genes was taken as a tag to show a strong correlation with plasmablast frequency (Spearman ρ = 0.71). These results indicate that the relative abundance of plasmablasts can be measured in whole blood samples using the three selected gene tags. Example 3: Correlation between plasmablast labeling and enhanced disease activity in SLE Previous work has shown a strong correlation between plasmablast frequency and disease severity, such as by estrogen safety in lupus erythematosus ( SELENA)-Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) score. This correlation appears to be driven by differences in patients with static and active disease. Observed from the Phase II clinical trial of EXPLORER [4] Moderate to severe extrarenal lupus erythematosus, and found that plasmablast cell label display has a lower but significant correlation with disease activity (Spearman ρ = 0.19, p = 0.03, Figure 3A). Looking more closely at the SLEDAI components driving this correlation, it was found that the three daughter scores in particular were particularly associated with high plasmablast abundance: DNA binding, low complement, and lymphocytopenia (Figure 3B-1, Figure 3B- 2. Figure 3B-3). A moderately negative correlation was found between plasmablast abundance and serum concentrations of complement components C3 and C4 in multiple lupus patient populations (Spilman ρ = -0.34, -0.38, respectively, Figure 3C). A moderate correlation was also observed between plasmablast content in these populations and the titer of anti-double-stranded DNA antibodies (Spearman ρ = 0.39, Figure 3C-1, Figure 3C-2, Figure 3C-3 ). Most patients with lupus display a transcriptional tag for interferon activity [2,5]. Plasmablast tags display a moderate correlation with interferon activity measured using three gene tags (Figures 3D-1 and 3D-2; [5]). The correlation appears to be driven by elevated plasmablast labeling in a subset of patients with high levels of interferon activity, with most of the lower interferon-labeled patients having lower plasmablast gene expression and high interferon activity Patients demonstrated a mix of lower plasmasomal gene expression and higher plasmasomal gene expression. However, plasmablast labeling is independent of interferon labeling and is related to disease severity and serological activity; using backward model selection, where the Akaike information criterion is used as a measure, plasmablasting and interferon labeling predicts serum complement content and plasmablastic cells Tag alone predicts anti-dsDNA antibody titer and SLEDAI. Whether as a combined score or for individual disease areas, no association was seen between plasmablast labeling scores and the British Isles Lupus Assessment Group (BILAG) activity index. These data support the role of plasmablasts in the activity of serological diseases driven by autoantibodies, lymphocytopenia, and hypocomplementemia. Example 4: Rituximab treatment reduces plasmablast cell label Plasmablast cells were collected from two moderate to severe lupus patient populations from a phase II clinical trial evaluating the safety and efficacy of rituximab in SLE The label value. These groups are patients with lupus nephritis (LUNAR) or extrarenal lupus (EXPLORER) [4,6]. The mixed effects model the plasmablast cell label value during the treatment course and incorporate the covariate age, race, combined medication, interferon activity, SLEDAI, consultation, treatment team and their interactions, where patients are modeled as random effects, Significant reductions in plasmablast labeling were identified in patients treated with rituximab (Figure 4A, Figure 4B). This effect was most apparent at the time point two weeks after receiving rituximab infusion, and the effect diminished over time. In the EXPLORER trial, two weeks after the fourth infusion of rituximab, a maximum reduction of 3.48 times was observed at week 28 (p = 2 × 10-11 ). Similarly, in the LUNAR test, the lowest content of plasmablast labeling was observed at the 28-week time point, with a 3.31-fold reduction (p = 0.0011). In the EXPLORER test, patients are monitored for the presence of antibodies to mouse human chimeric antibodies (HACA). Although most patients treated with rituximab showed a decrease in plasmablast labeling, patients who continued to show anti-drug antibodies showed no reduction in plasmablast labeling after rituximab treatment (Figure 4C) . As described above, using the same linear mixed effects model, plasmablast marker gene expression was found to be 2.8 times higher in patients who continued to develop HACA (p = 0.0007). Example 5: Standard Nursing Care for Lupus Changes Plasmablast Cell Labeling Previous studies have identified associations between different immunosuppressive therapies and reduced plasmablast-associated gene expression [7]. Observing the screening samples from the EXPLORER clinical trial, compared with patients treated with azathioprine, extremely low plasmablast labeling was found in patients treated with mycophenolate or methotrexate (Figure 5A). A trend toward decreased plasmablast gene expression in patients from the LUNAR trial was observed in patients treated with mycophenolate at baseline (Figure 5B), although relatively few patients were in the MMF treatment pool. Example 6: Patient Race / Ethnicity Affects Plasmablast Tag Content Biomarker content often varies between patient populations. Data analysis showed significantly lower plasmablast labeling in both the EXPLORER and ROSE clinical trial populations compared to patients of European descent with African or American Hispanic descent. This is true when considering interferon activity, age, and disease severity (Figures 6A, 6B, 6C). Example 7: Effect of BTK inhibition on plasmablast differentiation: Plasmablast differentiation: Isolate memory B cells from a healthy donor PBMC (Miltenyi memory B cell isolation kit). For plasmablast differentiation, subsequent studies were performed on cytokines, IL-2 (20 U / ml), IL-10 (50 ng / ml), IL-15 (10 ng / ml), IL-6 (50 ng / ml) ), IFNa (10 ng / ml) in the presence of a mixture, culture 1.5 × 10 ^ 5 / ml memory B cells, and use ODN2006 (TLR-9 ligand) 5 ug / ml or CD40L (3 ug / ml) Stimulate for 5 days. Plasmablast differentiation was performed in the presence of a separate vehicle (DMSO) and various concentrations of GDC-0852, and titration was performed using a three-fold dose of inhibitor starting at 10 uM. Perform flow cytometry to enumerate (CD20- CD38++ ) Percent of plasmablasts and evaluation of GDC-0852 inhibition. RNA preparation: RNA was extracted from a cell-containing culture at 5 days, which was derived from cells treated with DMSO and GDC-0852 (370 nM) (n = 4). Cells were disrupted in RLT buffer using Qiashredder (Qiagen, Valencia, CA), and RNA was subsequently extracted using an RNeasy mini kit (Qiagen) including on-column DNase digestion. NanoDrop 8000 (Thermo Scientific) was used to determine the total RNA concentration and the integrity of the RNA sample. The isolated RNA was used for Fluidigm quantitative RT-PCR analysis. QT PCR: cDNA synthesis was performed on 100 ng total-RNA using the iScript cDNA Synthesis Kit (Biorad, Hercules, CA). Gene-specific preamplification (Applied Biosystems) was performed on 3 genes (IgJ, MZB1, TXNDC5, including the housekeeping gene TMEM55B). RT-PCR was performed using a BioMark 48.48 dynamic array (Fluidigm Corporation) using the manufacturer's protocol. Data were collected using BioMark Data Collection Software and CT values were obtained using BioMark RT-PCR Analysis Software (V.2.1.1, Fluidigm). Calculate the relative abundance (dCt) of HPRT1: 2log- (mean Ct gene-mean Ct HPRT1). For statistical analysis, set the value below the lower detection limit to 1 Ct below the lowest recorded value. Perform statistical analysis using <> or custom scripts written in R programming language. To identify differences in gene expression, we fit a linear mixed-effects model to the relative transcriptional abundance of log2 transformations, with treatment as a fixed effect and donor as a random effect. To compare the percentage of plasmablasts between DMSO and compound-treated samples, we used the Wilcoxon rank sum test. The IC50 value of BTK inhibition was calculated using GraphPad Prism software. Human recombinant interleukin (IL) -2 and interferon-α (IFN-α) were purchased from R & D system (Minneapolis, MN) and IL-10, IL-6 and IL-15 were purchased from Peprotech (Rocky Hill, NJ) . CpG (ODN2006) was purchased from Invivogen (San Deigo, CA) and CD40L was purchased from R & D Systems (Minneapolis, MN).result : Differentiation of B cells into plasmablasts can occur via multiple activation stimuli and involves different molecular changes. CD20 is activated by CD40 and / or Toll like TLR+ CD27++ Memory B cells differentiate into CD20- CD38++ Plasmablast. We evaluated the effect of the BTK inhibitor GDC-0852 in the plasmablast differentiation method by using a CD40L-stimulated T cell-mediated response or a T cell-independent response using a Tuo receptor ligand CpG. GDC-0852 inhibited CD40L-induced plasmablast differentiation on day 5 in a dose-dependent manner, with an IC50 potency of 20.0 nM (+/- 0.002) (Figure 7). Gene expression analysis of cells treated with DMSO and GDC-0852 shows plasmablast cell tag gene IgJ(p = 0.011 ), MZB1 (p = 0.0023 ), TXNDC5 (p = 0.0032 ) And three plasma tags of compound plasmablasts (p = 0.0026 ) Significantly reduced, while native B cells displayed extremely low levels of tagged gene expression (p <1 × 10-6 ) (Figure 8). Comparing gene expression values with plasmablast abundance, we found a strong correlation between the three gene tags and the percentage of plasmablasts (Spilman ρ = 0.81) (Figure 9). CpG-mediated plasmablast differentiation (n = 3) was also inhibited by GDC-0852, with an IC50 potency of 48 nM (+/- 57) (Figure 10). When introducing elements of the invention or its preferred embodiments, the articles "a, an", "the" and "said" mean that one or more of the elements are present. The terms "comprising", "including" and "having" are intended to be inclusive and mean that there may be elements other than the listed elements. Although the foregoing invention has been described in considerable detail by way of illustration and examples for purposes of clear understanding, these descriptions and examples should not be construed as limiting the scope. The disclosures of all patents and scientific literature cited herein are expressly incorporated herein by reference in their entirety. References 1. Arce E, Jackson DG, Gill MA, Bennett LB, Banchereau J, Pascual V. Increased frequency of pre-germinal center B cells and plasma cell precursors in the blood of children with systemic lupus erythematosus. J Immunol. 2001; 167: 2361-2369. Doi: 10.4049 / jimmunol.167.4.2361 2. Bennett L, Palucka a K, Arce E, Cantrell V, Borvak J, Banchereau J, et al. Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med. 2003; 197: 711-723. Doi: 10.1084 / jem.20021553 3. Kalunian KC, Merrill JT, Maciuca R, McBride JM, Townsend MJ, Wei X, et al. A Phase II study of the efficacy and safety of rontalizumab (rhuMAb interferon-α) in patients with systemic lupus erythematosus (ROSE). Ann Rheum Dis. 2016; 75: 196-202. doi: 10.1136 / annrheumdis-2014-206090 4. Merrill JT, Neuwelt CM, Wallace DJ, Shanahan JC, Latinis KM, Oates JC, et al. Efficacy and safety of rituximab in moderately-to-severely active systemic lupus erythematosus: The randomized, double-blind, pha se II / III systemic lupus erythematosus evaluation of rituximab trial. Arthritis Rheum. 2010; 62: 222-233. doi: 10.1002 / art.27233 5. Kennedy WP, Maciuca R, Wolslegel K, Tew W, Abbas AR, Chaivorapol C, et al. Association of the interferon signature metric with serological disease manifestations but not global activity scores in multiple cohorts of patients with SLE. Lupus Sci Med. 2015; 2: e000080. doi: 10.1136 / lupus-2014-000080 6. Rovin BH, Furie R, Latinis K, Looney RJ, Fervenza FC, Sanchez-Guerrero J, et al. Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: the Lupus Nephritis Assessment with Rituximab study. Arthritis Rheum. 2012; 64: 1215- 1226. doi: 10.1002 / art.34359 7. Banchereau R, Hong S, Cantarel B, Baldwin N, Baisch J, Edens M, et al. Personalized Immunomonitoring Uncovers Molecular Networks that Stratify Lupus Patients. Cell. Elsevier Inc .; 2016; 165: 551-565. Doi: 10.1016 / j.cell.2016.03.008

專利或申請案文件含有至少一個彩製圖式。在申請且支付必要費用後,專利局將提供具有彩圖之此專利或專利申請公開案之複本。 1A-1 及圖 1A-2. 活體外漿母細胞分化及分類策略。在7天的含有CpG之培養條件下與細胞介素IL-2、IL-6、IL-10、IL-15、IFNα一起自CD20+CD27+記憶B細胞分化出漿母細胞。原生B細胞(CD20+CD27-)及經FACS分類之CD20+CD27+活化B細胞及經分化之CD20loCD38+漿母細胞用於基因表現分析。 1B. 由漿母細胞特異性地表現之基因之熱圖。鑑別出在0.001之FDR下,由漿母細胞比原生B細胞及活化B細胞高至少10倍表現且在漿母細胞中表現含量為>5 RPKM的基因。值表示方差穩定資料,其標準化為各基因內平均值0,標準差1。 2A. 其中添加有增加數目之漿母細胞的PBMC樣本中之候選漿母細胞標籤基因之熱圖。將漿母細胞自兩個單獨供體摻入至PBMC中,如熱圖上方之黑色及灰色中所指示。值表示各基因相對於HPRT1之ΔCt,且標準化為平均值0及標準差1。 2B-1 2B-2 2B -3 2B-4. 相較於存在於各樣本中之漿母細胞百分比,相對於HPRT1之漿母細胞標籤基因之表現含量,或全部三種基因之平均值。點線及短劃線指示不同PBMC供體,而不同符號表示漿母細胞之不同供體。線性回歸分析用於預測漿母細胞標籤或組分基因之表現,從而將PBMC供體及漿母細胞供體併入至模型中。全部四種模型為高度統計顯著的,其中p<1×10-10。該模型之預測能力報導為來自線性模型化之r2。 2C-1 2C-2 2C-3 2C-4. 漿母細胞基因對HPRT1之相對表現或接受流感痘苗一週後與健康供體分離之B細胞群體中所量測之全部三種標籤基因之平均值。N=原生B細胞,M=記憶B細胞,PB=漿母細胞。相較於其他群體,漿母細胞具有最高之標記基因表現。星形指示B細胞群體之間的差異之統計顯著性,使用線性回歸,包括供體作為共變數;*=p<0.05,**=p<0.01,***=p<0.001。 2D-1 2D-2 2D-3 2D-4. 漿母細胞標籤及組分基因與由FACS在狼瘡患者血液中量測之漿母細胞之頻率相關。對於總共96個樣本,在多達3個時間點內,IgD-CD19+CD27++CD38++漿母細胞經量測為43名患者中之全血細胞百分比,對於其吾等隨附RNA定序資料。基因表現值表示為個別基因之RPKM或三種基因標籤之幾何平均RPKM。使用斯皮爾曼等級次序方法(Spearman's rank-order method)計算相關係數。 3A . 漿母細胞標籤與由SLEDAI量測之疾病活動性相關。值表示漿母細胞標籤基因相對於HPRT1之平均表現。SLEDAI及漿母細胞平均表現值來自開始治療前所收集的樣本。使用斯皮爾曼等級次序方法測定相關係數。 3B-1 3B-2 3B-3. SLEDAI組合物指數之個別組分與增加之漿母細胞標籤基因表現相關聯。線性回歸用於分析展現症狀中之每一者之患者與未展現症狀之患者之間的統計顯著性;星形指示此測試中之顯著性含量:*=p<0.05,**=p<0.01,***=p<0.001。 3C-1 3C-2 3C-3. 血清C3及C4補體含量及血清抗dsDNA抗體效價與漿母細胞標籤表現相關。使用斯皮爾曼等級次序方法計算相關係數。 3D-1 3D-2. 全血干擾素標籤表現(ISM)與漿母細胞標籤值相關。使用斯皮爾曼等級次序方法計算相關係數。 4A . 用利妥昔單抗治療患者減少漿母細胞標籤表現含量。線指示利妥昔單抗治療群(虛線)或安慰劑群(實線)內之平均表現含量,其中誤差條指示平均值之標準誤差。黑色箭頭指示患者何時接受輸注藥物或安慰劑。漿母細胞標籤基因之表現使用線形混合效應模型建模,併入年齡、人種/種族、所使用之合併用藥、干擾素活性、SLEDAI及治療隊組及時間點及其作為固定效應之相互作用以及作為隨機效應之患者。紅色星形指示特定而言利妥昔單抗治療組中顯著不同的時間點:*=p<0.05,**=p<0.01,***=p<0.001。 4B . 分別用黴酚酸酯及利妥昔單抗治療減少漿母細胞標籤表現含量。線指示安慰劑群(實線)或利妥昔單抗治療群(虛線)之平均值,其中平均值之標準誤差由誤差條指示。箭頭指示患者何時接受輸注安慰劑或利妥昔單抗。表現值使用線形混合效應模型建模,併入年齡、干擾素活性及治療隊組及問診及其相互作用,以及作為隨機效應之患者。曲線圖頂部處之星形指示其中經利妥昔單抗治療之患者展示超過安慰劑組之自基線顯著減少的時間點,而曲線圖之底部附近之星形指示無關於治療之不同於基線的時間點:*=p<0.05,**=p<0.01,***=p<0.001。 4C. 具有可偵測抗嵌合抗體(HACA)之患者具有較高漿母細胞標記基因表現。線指示具有可偵測HACA之經利妥昔單抗治療之患者(實線)或從未具有可偵測HACA之彼等患者(虛線)中之漿母細胞標記基因之平均表現,誤差條指示平均值之標準誤差。 5A . 用黴酚酸嗎啉乙酯(MMF)或甲胺喋呤(MTX)治療之患者展示比用硫唑嘌呤(AZA)治療之患者更低之漿母細胞表現。在進行不同免疫抑制方案之患者之間比較篩檢漿母細胞標籤值。使用線性回歸測試統計顯著性,將AZA與其他兩種治療中之每一者進行比較。星形指示治療之間的顯著差異:*=p<0.05,**=p<0.01,***=p<0.001。 5B. 在篩檢時進行MMF治療之患者傾向於具有比未進行MMF治療之患者更低之漿母細胞標籤。在採用MMF之患者與未採用MMF之彼等患者之間在其之篩檢問診時使用線性回歸計算p值。 6A . 在EXPLORER臨床試驗群中,具有歐洲血統之患者具有比其他種族更低之漿母細胞表現含量。值表示漿母細胞基因相對於HPRT1之平均表現含量。使用線性回歸在自報導人種/種族中比較篩檢問診值。星形指示相較於自報導為白人/白種人之患者的差異顯著性:*=p<0.05,**=p<0.01,***=p<0.001。 6B . LUNAR患者展示基於人種/種族之無顯著差異。使用線性回歸,未在種族中觀測到顯著差異。 6C . 在ROSE臨床試驗群中具有歐洲血統之患者展示較低之漿母細胞標記表現。值表示漿母細胞基因之幾何平均RPKM。使用線性回歸針對log2轉化平均RPKM在自報導人種/種族中比較篩檢問診值。星形指示相較於自報導為白人/白種人之患者的差異顯著性:*=p<0.05,**=p<0.01,***=p<0.001。 7 .藉由BTK抑制劑GDC-0852抑制CD40L介導之漿母細胞分化的劑量反應曲線展示以劑量依賴方式抑制漿母細胞分化。使用FACS分析來測定四個健康供體中之漿母細胞百分比以計算各供體之IC50值。 8A 8B 8C 8D. BTK抑制減少漿母細胞基因標籤。在DMSO媒劑或370nM GDC-0852之存在下,使用上文所描述的條件將來自4個健康供體之記憶B細胞分化成漿母細胞。藉由Fluidigm來量測漿母細胞標籤基因之表現,且標準化為管家基因(HPRT1)。表現值經繪製為管家基因之相對轉錄豐度。漿母細胞標籤經計算為三種個別基因之相對豐度的幾何平均值。 9 . 漿母細胞基因表現與漿母細胞細胞數目相關。如由FACS分析所測定之漿母細胞百分比與漿母細胞標籤相關,如圖8D中所測定(斯皮爾曼ρ=0.81)。白點指示在DMSO之存在分化之樣本,而黑點表示經GDC-0852治療之樣本。 10 . BTK抑制劑GDC-0852以劑量依賴方式抑制CpG介導之漿母細胞分化。使用FACS分析來測定四個健康供體中之漿母細胞百分比以計算各供體之IC50值。The patent or application file contains at least one color drawing. After applying for and paying the necessary fees, the Patent Office will provide a copy of this patent or patent application publication with color graphics. Figure 1A-1 and Figure 1A-2. In vitro plasmablast differentiation and classification strategies. Plasmablasts were differentiated from CD20 + CD27 + memory B cells together with cytokines IL-2, IL-6, IL-10, IL-15, and IFNα under 7-day culture conditions containing CpG. Native B cells (CD20 + CD27-) and CD20 + CD27 + activated B cells classified by FACS and differentiated CD20loCD38 + plasmablasts were used for gene expression analysis. Figure IB. Heat map of genes specifically expressed by plasmablasts. Under the FDR of 0.001, genes with at least 10-fold higher expression in plasmablasts than in native B cells and activated B cells and plasma cells with a content> 5 RPKM were identified. Values represent stable-variance data, which are normalized to a mean of 0 within each gene and a standard deviation of 1. Figure 2A. Heat map of candidate plasmablast tag genes in PBMC samples to which an increased number of plasmablasts are added. Plasmablasts were incorporated into PBMC from two separate donors, as indicated in black and gray above the heat map. Values represent the ΔCt of each gene relative to HPRT1 and are normalized to mean 0 and standard deviation 1. Figure 2B-1 , 2B-2 , 2B - 3, and 2B-4. Compared with the percentage of plasmablasts present in each sample, relative to the expression content of plasmablast tag genes in HPRT1, or the average of all three genes value. Dotted and dashed lines indicate different PBMC donors, and different symbols indicate different donors of plasmablasts. Linear regression analysis is used to predict the performance of plasmablast cell tags or component genes, thereby incorporating PBMC donors and plasmablast donors into the model. All four models are highly statistically significant, with p <1 × 10-10. The predictive power of this model is reported as r2 from linear modeling. Figure 2C-1 , 2C-2 , 2C-3, and 2C-4. Relative performance of plasmablast genes to HPRT1 or all three tag genes measured in a B-cell population isolated from a healthy donor after receiving influenza vaccination one week The average. N = native B cells, M = memory B cells, PB = plasmablasts. Compared with other populations, plasmablasts have the highest marker gene performance. Stars indicate statistical significance of differences between B cell populations, using linear regression including donors as covariates; * = p <0.05, ** = p <0.01, *** = p <0.001. Figures 2D-1 , 2D-2 , 2D-3, and 2D-4. Plasmablast cell tags and component genes correlate with the frequency of plasmablast cells measured by FACS in the blood of patients with lupus. For a total of 96 samples, IgD-CD19 + CD27 ++ CD38 ++ plasmablasts were measured as the percentage of whole blood cells in 43 patients for up to 3 time points, and for our accompanying RNA sequencing data . Gene expression values are expressed as the RPKM of individual genes or the geometric mean RPKM of three gene tags. The correlation coefficient was calculated using Spearman's rank-order method. Figure 3A . Plasmablast labeling correlates with disease activity measured by SLEDAI. Values represent the average performance of plasmablast tag genes relative to HPRT1. The average SLEDAI and plasmablast performance values were obtained from samples collected before starting treatment. The correlation coefficient was determined using the Spearman ranking method. Figures 3B-1 , 3B-2, and 3B-3. Individual components of the SLEDAI composition index are correlated with increased plasmablast tag gene expression. Linear regression was used to analyze the statistical significance between patients who exhibited each of the symptoms and patients who did not exhibit symptoms; a star indicates the significance content in this test: * = p <0.05, ** = p <0.01 , *** = p <0.001. Figures 3C-1 , 3C-2, and 3C-3. Serum C3 and C4 complement levels and serum anti-dsDNA antibody titers correlate with plasmablast labeling performance. The correlation coefficient was calculated using the Spearman ranking method. Figures 3D-1 and 3D-2. The whole blood interferon labeling performance (ISM) correlates with plasmablast labeling. The correlation coefficient was calculated using the Spearman ranking method. Figure 4A . Treatment of patients with rituximab reduces plasmablast labeling content. Lines indicate average performance levels within the rituximab treatment group (dashed line) or placebo group (solid line), where error bars indicate the standard error of the mean. Black arrows indicate when the patient received an infusion of medication or placebo. The performance of plasmablast tag genes was modeled using a linear mixed-effects model, incorporating age, ethnicity / ethnicity, combined medications used, interferon activity, SLEDAI, and treatment team and time points and their interactions as fixed effects As well as patients with random effects. Red stars indicate significantly different time points in the rituximab-treated group in particular: * = p <0.05, ** = p <0.01, *** = p <0.001. Figure 4B . Treatment with mycophenolate mofetil and rituximab, respectively, reduces plasmablast labeling content. Lines indicate the mean of the placebo group (solid line) or the rituximab treatment group (dashed line), where the standard error of the mean is indicated by the error bar. Arrows indicate when the patient received an infusion of placebo or rituximab. Performance values were modeled using a linear mixed effects model, incorporating age, interferon activity, and treatment team and interrogation and their interactions, as well as patients with random effects. The star at the top of the graph indicates the time point in which patients treated with rituximab showed a significant decrease from baseline over the placebo group, while the star near the bottom of the graph indicates that there is no Time points: * = p <0.05, ** = p <0.01, *** = p <0.001. Figure 4C. Patients with detectable anti-chimeric antibodies (HACA) have higher plasmablast gene expression. Lines indicate the average performance of plasmablast marker genes in patients treated with rituximab that detect HACA (solid line) or those that have never detected HACA (dotted line). Error bars indicate Standard error of the mean. Figure 5A . Patients treated with mycophenolate morpholinate (MMF) or methotrexate (MTX) demonstrated lower plasmablast performance than patients treated with azathioprine (AZA). Screening plasmablast label values among patients undergoing different immunosuppressive regimens. Linear regression tests were used for statistical significance to compare AZA with each of the other two treatments. Stars indicate significant differences between treatments: * = p <0.05, ** = p <0.01, *** = p <0.001. Figure 5B. Patients treated with MMF at screening tend to have a lower plasmablast label than patients not treated with MMF. Linear regression was used to calculate p-values between patients with MMF and those without MMF at their screening visits. Figure 6A . In the EXPLORER clinical trial group, patients with European ancestry had lower plasmablast performance than other races. Values represent the mean expressed content of plasmablast genes relative to HPRT1. Use linear regression to compare screening questionnaire values among self-reported races / races. The star-shaped indications are significant compared to patients who have reported themselves as white / white: * = p <0.05, ** = p <0.01, *** = p <0.001. Figure 6B . LUNAR patients show no significant differences based on race / ethnicity. Using linear regression, no significant differences were observed in the race. Figure 6C . Patients with European ancestry in the ROSE clinical trial population show lower plasmablast performance. Values represent the geometric mean RPKM of plasmablast genes. Screening diagnostic values were compared among self-reported races / races using linear regression for log2 transformed average RPKM. The star-shaped indications are significant compared to patients who have reported themselves as white / white: * = p <0.05, ** = p <0.01, *** = p <0.001. Figure 7. Dose-response curve of CD40L-mediated plasmablast differentiation with BTK inhibitor GDC-0852 showing inhibition of plasmablast differentiation in a dose-dependent manner. FACS analysis was used to determine the percentage of plasmablasts in four healthy donors to calculate the IC50 value for each donor. Figures 8A , 8B , 8C and 8D. BTK inhibition reduces plasmablast gene tag. In the presence of DMSO vehicle or 370nM GDC-0852, memory B cells from 4 healthy donors were differentiated into plasmablasts using the conditions described above. The performance of plasmablast cell tag genes was measured by Fluidigm and standardized as the housekeeping gene (HPRT1). The performance values are plotted as the relative transcriptional abundance of the housekeeping gene. The plasmablast label was calculated as the geometric mean of the relative abundances of the three individual genes. FIG. Plasmablast gene expression related to the number of parent cells cytoplasm. The percentage of plasmablasts as determined by FACS analysis correlates with plasmablast labeling, as determined in Figure 8D (Spearman ρ = 0.81). White dots indicate the presence of differentiated samples in DMSO, while black dots indicate the samples treated with GDC-0852. FIG 10. BTK inhibitor GDC-0852 in a dose-dependent inhibition of plasma blasts manner CpG-mediated differentiation. FACS analysis was used to determine the percentage of plasmablasts in four healthy donors to calculate the IC50 value for each donor.

Claims (19)

一種BTK抑制劑在製造用於治療患有自體免疫或發炎疾病之個體之藥物中之用途,其中已發現來自該個體之樣本具有升高含量之選自由IgJ、Mzb1及Txndc5組成之群的一或多種生物標記物。Use of a BTK inhibitor in the manufacture of a medicament for the treatment of an individual suffering from an autoimmune or inflammatory disease, in which samples from the individual have been found to have an increased content of one selected from the group consisting of IgJ, Mzb1 and Txndc5 Or multiple biomarkers. 一種為患有自體免疫或發炎疾病之個體選擇療法的方法,其包含測定選自由IgJ、Mzb1及Txndc5組成之群的一或多種生物標記物之含量;以及基於該等生物標記物之含量來選擇藥物。A method of selecting a therapy for an individual suffering from an autoimmune or inflammatory disease, comprising determining the content of one or more biomarkers selected from the group consisting of IgJ, Mzb1 and Txndc5; and selecting based on the content of the biomarkers drug. 一種鑑別患有自體免疫或發炎疾病之個體是否可能展現受益於包含BTK抑制劑之治療的方法,其係測定來自該個體之樣本中之選自由IgJ、Mzb1及Txndc5組成之群的一或多種生物標記物之含量,其中該樣本中之該等生物標記物含量升高指示該個體很可能展現受益於包含該BTK抑制劑之治療,或該等生物標記物含量降低指示該個體不大可能展現受益於包含該BTK抑制劑之治療。A method of identifying whether an individual with an autoimmune or inflammatory disease is likely to exhibit benefit from a treatment comprising a BTK inhibitor, which comprises determining one or more of a sample from the individual selected from the group consisting of IgJ, Mzb1, and Txndc5 Biomarker content, where an increase in the biomarker content in the sample indicates that the individual is likely to exhibit benefit from treatment comprising the BTK inhibitor, or a decrease in the biomarker content indicates that the individual is unlikely to exhibit Benefit from treatments that include this BTK inhibitor. 一種鑑別患有自體免疫或發炎疾病之個體接受BTK抑制劑之分析法,該方法包含: (a)測定來自該個體之樣本中之選自由IgJ、Mzb1及Txndc5組成之群的一或多種生物標記物之含量;以及 (b)基於該等生物標記物之該等含量推薦投與BTK抑制劑。An assay for identifying an individual with an autoimmune or inflammatory disease as receiving a BTK inhibitor, the method comprising: (a) determining one or more organisms selected from the group consisting of IgJ, Mzb1, and Txndc5 in a sample from the individual The content of the marker; and (b) it is recommended to administer a BTK inhibitor based on the content of the biomarkers. 一種包含一或多種反應劑之診斷套組,其用於測定來自患有自體免疫或發炎疾病之個體之樣本中的選自由IgJ、Mzb1及Txndc5組成之群之一或多種生物標記物的含量,其中偵測到升高含量之該等生物標記物意謂當用BTK抑制劑治療該個體時療效提高,且其中偵測到較低或實質上無法偵測到含量之生物標記物意謂當用BTK抑制劑治療患有該自體免疫或發炎疾病之個體時療效降低。A diagnostic kit comprising one or more reactants for determining the content of one or more biomarkers selected from the group consisting of IgJ, Mzb1 and Txndc5 in a sample from an individual suffering from an autoimmune or inflammatory disease Where the detection of elevated levels of these biomarkers means that the efficacy is improved when the individual is treated with a BTK inhibitor, and where the detection of lower or substantially undetectable levels of biomarkers means when The use of BTK inhibitors in treating individuals suffering from this autoimmune or inflammatory disease has reduced efficacy. 如請求項2及3中任一項之方法,其中該方法進一步包含向該個體投與有效量之BTK抑制劑。The method of any one of claims 2 and 3, wherein the method further comprises administering to the individual an effective amount of a BTK inhibitor. 如請求項1之用途、如請求項2或3之方法、如請求項4之分析法、或如請求項5之套組,其中生物標記物含量之測定法係量測該等生物標記物之RNA含量,與參考含量進行相對比較。If the purpose of claim 1, the method of claim 2 or 3, the analysis method of claim 4, or the set of claim 5, the method of measuring the content of biomarkers is to measure the biomarker content. Relative RNA content compared to reference content. 如請求項7之用途、方法、分析法或套組,其中量測該等RNA含量包括擴增。If the use, method, analysis, or kit of claim 7, wherein measuring the RNA content includes amplification. 如請求項8之用途、方法、分析法或套組,其中量測該等RNA含量包括定量PCR。If the use, method, analysis, or set of claim 8, the measurement of the RNA content includes quantitative PCR. 如請求項9之用途、方法、分析法或套組,其中量測該等RNA含量包括擴增RNA且偵測擴增產物,由此量測該RNA相對於參考含量之含量。If the use, method, analysis method or kit of claim 9, wherein measuring the content of the RNA includes amplifying the RNA and detecting the amplification product, the content of the RNA relative to the reference content is measured. 如請求項1之用途、如請求項2或3之方法、如請求項4之分析法、或如請求項5之套組,其中該樣本為血液樣本。If the purpose of claim 1, the method of claim 2 or 3, the analysis of claim 4, or the kit of claim 5, wherein the sample is a blood sample. 如請求項1之用途、如請求項2或3之方法、如請求項4之分析法、或如請求項5之套組,其中該BTK抑制劑為抗體、結合多肽、小分子及/或聚核苷酸。If the use of claim 1, the method of claim 2 or 3, the analysis of claim 4, or the set of claim 5, wherein the BTK inhibitor is an antibody, binding polypeptide, small molecule and / or polymer Nucleotides. 如請求項12之用途、方法、分析法或套組,其中該BTK抑制劑為小分子。If the use, method, analysis or kit of claim 12, wherein the BTK inhibitor is a small molecule. 如請求項13之用途、方法、分析法或套組,其中該小分子為化合物(A):或其醫藥學上可接受之鹽。If the use, method, analysis or set of claim 13, wherein the small molecule is compound (A): Or a pharmaceutically acceptable salt thereof. 如請求項2及3中任一項之用途、方法、分析法或套組,其中該自體免疫或發炎疾病為全身性紅斑性狼瘡症。The use, method, analysis or kit of any one of claims 2 and 3, wherein the autoimmune or inflammatory disease is systemic lupus erythematosus. 如請求項15之用途、方法、分析法或套組,其中該自體免疫或發炎疾病為狼瘡性腎炎。The use, method, analysis or kit of claim 15, wherein the autoimmune or inflammatory disease is lupus nephritis. 如請求項15之用途、方法、分析法或套組,其中該自體免疫或發炎疾病為腎外狼瘡。The use, method, analysis or kit of claim 15, wherein the autoimmune or inflammatory disease is extrarenal lupus. 如請求項2及3中任一項之用途、方法、分析法或套組,其中選擇該等生物標記物中之兩者。If the use, method, analysis or kit of any one of claims 2 and 3 is selected, both of the biomarkers are selected. 如請求項2及3中任一項之用途、方法、分析法或套組,其中選擇該等生物標記物中之三者。If the use, method, analysis or set of any one of claims 2 and 3 is selected, three of these biomarkers are selected.
TW107110152A 2017-03-24 2018-03-23 Methods of treating autoimmune and inflammatory diseases TW201836642A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762476406P 2017-03-24 2017-03-24
US62/476,406 2017-03-24

Publications (1)

Publication Number Publication Date
TW201836642A true TW201836642A (en) 2018-10-16

Family

ID=62063588

Family Applications (1)

Application Number Title Priority Date Filing Date
TW107110152A TW201836642A (en) 2017-03-24 2018-03-23 Methods of treating autoimmune and inflammatory diseases

Country Status (6)

Country Link
US (1) US20200124600A1 (en)
EP (1) EP3602058A1 (en)
JP (1) JP2020514384A (en)
CN (1) CN110709702A (en)
TW (1) TW201836642A (en)
WO (1) WO2018175863A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI698640B (en) * 2018-04-23 2020-07-11 長庚醫療財團法人高雄長庚紀念醫院 Methods for detecting or assessing the risk of developing lupus nephritis and application thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3914250A1 (en) 2019-01-22 2021-12-01 F. Hoffmann-La Roche AG Methods of treating rheumatoid arthritis, chronic spontaneous urticaria, and systemic lupus erythematosis using an inhibitor of bruton's tyrosine kinase

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4018653A (en) 1971-10-29 1977-04-19 U.S. Packaging Corporation Instrument for the detection of Neisseria gonorrhoeae without culture
US4016043A (en) 1975-09-04 1977-04-05 Akzona Incorporated Enzymatic immunological method for the determination of antigens and antibodies
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4318980A (en) 1978-04-10 1982-03-09 Miles Laboratories, Inc. Heterogenous specific binding assay employing a cycling reactant as label
US4424279A (en) 1982-08-12 1984-01-03 Quidel Rapid plunger immunoassay method and apparatus
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
US6335155B1 (en) 1998-06-26 2002-01-01 Sunesis Pharmaceuticals, Inc. Methods for rapidly identifying small organic molecule ligands for binding to biological target molecules
WO2000039585A1 (en) 1998-12-28 2000-07-06 Sunesis Pharmaceuticals, Inc. Identifying small organic molecule ligands for binding
US7405295B2 (en) 2003-06-04 2008-07-29 Cgi Pharmaceuticals, Inc. Certain imidazo[1,2-a]pyrazin-8-ylamines and method of inhibition of Bruton's tyrosine kinase by such compounds
WO2005005429A1 (en) 2003-06-30 2005-01-20 Cellular Genomics, Inc. Certain heterocyclic substituted imidazo[1,2-a]pyrazin-8-ylamines and methods of inhibition of bruton’s tyrosine kinase by such compounds
EP1812442A2 (en) 2004-11-10 2007-08-01 CGI Pharmaceuticals, Inc. Imidazo[1,2-a]pyrazin-8-ylamines useful as modulators of kinase activity
BRPI0608252A2 (en) 2005-03-10 2010-04-06 Cgi Pharmaceuticals Inc chemical entities, pharmaceutical compositions comprising them, methods using said chemical entities and use of said chemical entities
US7838523B2 (en) 2006-09-11 2010-11-23 Cgi Pharmaceuticals, Inc. Certain substituted amides, method of making, and method of use thereof
AR063706A1 (en) 2006-09-11 2009-02-11 Cgi Pharmaceuticals Inc CERTAIN AMIDAS REPLACED, THE USE OF THE SAME FOR THE TREATMENT OF DISEASES MEDIATED BY THE INHIBITION OF THE ACTIVITY OF BTK AND PHARMACEUTICAL COMPOSITIONS THAT UNDERSTAND THEM.
AR063946A1 (en) 2006-09-11 2009-03-04 Cgi Pharmaceuticals Inc CERTAIN REPLACED PIRIMIDINS, THE USE OF THE SAME FOR THE TREATMENT OF DISEASES MEDIATED BY THE INHIBITION OF THE ACTIVITY OF BTK AND PHARMACEUTICAL COMPOSITIONS THAT UNDERSTAND THEM.
CL2008002793A1 (en) 2007-09-20 2009-09-04 Cgi Pharmaceuticals Inc Compounds derived from substituted amides, inhibitors of btk activity; pharmaceutical composition comprising them; Useful in the treatment of cancer, bone disorders, autoimmune diseases, among others
EP2107068A1 (en) * 2008-03-31 2009-10-07 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. MZB1, a novel B cell factor, and uses thereof
ES2554615T3 (en) 2008-05-06 2015-12-22 Gilead Connecticut, Inc. Substituted amides, method of preparation and use as Btk inhibitors
US8598174B2 (en) 2008-11-12 2013-12-03 Genetech, Inc. Pyridazinones, method of making, and method of use thereof
KR20140022815A (en) 2011-02-28 2014-02-25 제넨테크, 인크. Biological markers and methods for predicting response to b-cell antagonists
UA111756C2 (en) 2011-11-03 2016-06-10 Ф. Хоффманн-Ля Рош Аг HETEROARYLPYRIDONE AND AZAPIRIDONE COMPOUNDS AS BRUTON TYROSINKINASE INHIBITORS
US10323013B2 (en) * 2013-04-23 2019-06-18 H. Lee Moffitt Cancer Center And Research Institute, Inc. Inhibitors of the IRE-1/XBP-1 pathway and methods of using thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI698640B (en) * 2018-04-23 2020-07-11 長庚醫療財團法人高雄長庚紀念醫院 Methods for detecting or assessing the risk of developing lupus nephritis and application thereof

Also Published As

Publication number Publication date
EP3602058A1 (en) 2020-02-05
US20200124600A1 (en) 2020-04-23
JP2020514384A (en) 2020-05-21
CN110709702A (en) 2020-01-17
WO2018175863A1 (en) 2018-09-27

Similar Documents

Publication Publication Date Title
Raterman et al. The interferon type I signature towards prediction of non-response to rituximab in rheumatoid arthritis patients
US20220136057A9 (en) Methods of identifying responses to map kinase inhibition therapy
JP2015526078A (en) Markers associated with human double microchromosome 2 inhibitors
RU2600026C2 (en) Prognostic factors for treating cancer
US11913074B2 (en) Methods for assessing risk of developing a viral disease using a genetic test
US11946053B2 (en) Methods for overcoming glucocorticoid resistance and for determining glucocorticoid resistance potential in cancer
US20190192488A1 (en) Use of mitochondrial activity inhibitors for the treatment of poor prognosis acute myeloid leukemia
KR20190082247A (en) How to treat cancer patients using parnesyl transferase inhibitor
US20200124600A1 (en) Methods of treating autoimmune and inflammatory diseases
US20210186982A1 (en) Methods and compositions for treating melanoma
WO2018046615A1 (en) Methods and compositions for predicting chronic lung allograft dysfunction
CN113645991A (en) Methods, systems, and kits targeting IL18R1 for the treatment of inflammatory diseases
CA2697887A1 (en) Modulating and/or detecting activation induced deaminase and methods of use thereof
JP2010004750A (en) Method for diagnosing onset or onset possibility of tissue disorder by autoimmune disease, and utilization thereof
EP3487875A1 (en) Biomarkers for detection and treatment of mast cell activity-associated disorders
US11479817B2 (en) Methods for discriminating a tolerant subject
AU2013217940A1 (en) Methods related to treatment of inflammatory diseases and disorders
JP2018518198A (en) Prognostic method for lymphatic blood disease
TW202342766A (en) Precision therapy for the treatment of cancer
WO2021250546A1 (en) Micrornas as predictors of response to anti-ige therapies in chronic spontaneous urticaria
US9732320B2 (en) Selective differentiation, identification, and modulation of human TH17 cells
US20210251980A1 (en) Crenolanib for treating flt3 mutated proliferative disorders associated mutations
WO2023230610A2 (en) Biomarkers for chronic fatigue syndrome and long covid and uses thereof
Raterman et al. Pharmacogenomics in rituximab treated rheumatoid arthritis patients